EP3903828A1 - Combination of antibody-drug conjugate and kinase inhibitor - Google Patents

Combination of antibody-drug conjugate and kinase inhibitor Download PDF

Info

Publication number
EP3903828A1
EP3903828A1 EP19899188.7A EP19899188A EP3903828A1 EP 3903828 A1 EP3903828 A1 EP 3903828A1 EP 19899188 A EP19899188 A EP 19899188A EP 3903828 A1 EP3903828 A1 EP 3903828A1
Authority
EP
European Patent Office
Prior art keywords
inhibitor
antibody
pharmaceutical composition
amino acid
composition according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19899188.7A
Other languages
German (de)
French (fr)
Other versions
EP3903828A4 (en
Inventor
Chiaki Ishii
Yasuki Kamai
Kiyoshi Sugihara
Daisuke Okajima
Yuuri HASHIMOTO
Hirokazu Suzuki
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Daiichi Sankyo Co Ltd
Original Assignee
Daiichi Sankyo Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Daiichi Sankyo Co Ltd filed Critical Daiichi Sankyo Co Ltd
Publication of EP3903828A1 publication Critical patent/EP3903828A1/en
Publication of EP3903828A4 publication Critical patent/EP3903828A4/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68037Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a camptothecin [CPT] or derivatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6889Conjugates wherein the antibody being the modifying agent and wherein the linker, binder or spacer confers particular properties to the conjugates, e.g. peptidic enzyme-labile linkers or acid-labile linkers, providing for an acid-labile immuno conjugate wherein the drug may be released from its antibody conjugated part in an acidic, e.g. tumoural or environment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to a pharmaceutical composition wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination, and/or a method of treatment wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination to a subject.
  • kinase inhibitors are agents that inhibit kinases involved in abnormally activated intracellular signaling to exert an antitumor effect.
  • kinase inhibitors include a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, an PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, and an NTRK inhibitor (Non-Patent References
  • An antibody-drug conjugate having a drug with cytotoxicity conjugated to an antibody, whose antigen is expressed on the surface of cancer cells and which also binds to an antigen capable of cellular internalization, and therefore can deliver the drug selectively to cancer cells, is thus expected to cause accumulation of the drug within cancer cells and to kill the cancer cells (Non-Patent References 12 to 16).
  • an antibody-drug conjugate comprising an antibody and a derivative of exatecan, which is a topoisomerase I inhibitor, as its components is known (Patent References 1 to 8, Non-Patent References 17 to 21).
  • Patent References 1 to 8 disclose that the antibody-drug conjugate can be administered together with various cancer treating agents.
  • An antibody-drug conjugate used in the present invention (an antibody-drug conjugate including a derivative of exatecan as a component) has been confirmed to exert a superior antitumor effect even as a single agent.
  • an antibody-drug conjugate including a derivative of exatecan as a component has been confirmed to exert a superior antitumor effect even as a single agent.
  • An object of the present invention is to provide a pharmaceutical composition wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination, and/or a method of treatment wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination to a subject.
  • the present invention provides the following [1] to [1072].
  • the present invention can provide a pharmaceutical composition wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination, and/or a method of treatment wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination to a subject.
  • the antibody-drug conjugate used in the present invention is an antibody-drug conjugate in which a drug-linker represented by the following formula:
  • the partial structure consisting of a linker and a drug in the antibody-drug conjugate is referred to as a "drug-linker".
  • the drug-linker is connected to a thiol group (in other words, the sulfur atom of a cysteine residue) formed at an interchain disulfide bond site (two sites between heavy chains, and two sites between a heavy chain and a light chain) in the antibody.
  • the drug-linker of the present invention includes exatecan (IUPAC name: (1S,9S)-1-amino-9-ethyl-5-fluoro-1,2,3,9,12,15-hexahydro-9-hydroxy-4-methyl-10H,13H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-10,13-dione, (also expressed as chemical name: (1S,9S)-1-amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-10,13(9H,15H)-dione)), which is a topoisomerase I inhibitor, as a component.
  • Exatecan is a camptothecin derivative having an antitumor effect, represented by the following formula:
  • the antibody-drug conjugate used in the present invention can be also represented by the following formula: wherein, the drug-linker is conjugated to an antibody via a thioether bond.
  • the meaning of n is the same as that of what is called the average number of conjugated drug molecules (DAR; Drug-to-Antibody Ratio), and indicates the average number of units of the drug-linker conjugated per antibody molecule.
  • the antibody-drug conjugate used in the present invention is cleaved at the linker portion to release the compound represented by the following formula.
  • the aforementioned compound is inferred to be the original source of the antitumor activity of the antibody-drug conjugate used in the present invention, and has been confirmed to have a topoisomerase I inhibitory effect ( Ogitani Y. et al., Clinical Cancer Research, 2016, Oct 15;22(20):5097-5108, Epub 2016 Mar 29 ) .
  • the antibody-drug conjugate used in the present invention is known to have a bystander effect ( Ogitani Y. et al., Cancer Science (2016) 107, 1039-1046 ).
  • the bystander effect is exerted through a process such that the antibody-drug conjugate used in the present invention is internalized in cancer cells expressing a target and the aforementioned compound is released then exerts an antitumor effect also on cancer cells which are present therearound and not expressing the target.
  • This bystander effect is exerted as an excellent antitumor effect even when the antibody-drug conjugate is used in combination with a kinase inhibitor according to the present invention.
  • the antibody in the antibody-drug conjugate used in the present invention may be derived from any species, and is preferably an antibody derived from a human, a rat, a mouse, or a rabbit. In cases when the antibody is derived from species other than human species, it is preferably chimerized or humanized using a well known technique.
  • the antibody of the present invention may be a polyclonal antibody or a monoclonal antibody and is preferably a monoclonal antibody.
  • the antibody in the antibody-drug conjugate used in the present invention is an antibody preferably having a characteristic of being capable of targeting cancer cells, and is preferably an antibody possessing, for example, a property of recognizing a cancer cell, a property of binding to a cancer cell, a property of internalizing in a cancer cell, and/or cytocidal activity against cancer cells.
  • the binding activity of the antibody against cancer cells can be confirmed using flow cytometry.
  • the internalization of the antibody into cancer cells can be confirmed using (1) an assay of visualizing an antibody incorporated in cells under a fluorescence microscope using a secondary antibody (fluorescently labeled) binding to the therapeutic antibody ( Cell Death and Differentiation (2008) 15, 751-761 ), (2) an assay of measuring a fluorescence intensity incorporated in cells using a secondary antibody (fluorescently labeled) binding to the therapeutic antibody ( Molecular Biology of the Cell, Vol.
  • a Mab-ZAP assay using an immunotoxin binding to the therapeutic antibody wherein the toxin is released upon incorporation into cells to inhibit cell growth Bio Techniques 28: 162-165, January 2000 .
  • the immunotoxin a recombinant complex protein of a diphtheria toxin catalytic domain and protein G may be used.
  • the antitumor activity of the antibody can be confirmed in vitro by determining inhibitory activity against cell growth.
  • a cancer cell line overexpressing a target protein for the antibody is cultured, and the antibody is added at varying concentrations into the culture system to determine inhibitory activity against focus formation, colony formation, and spheroid growth.
  • the antitumor activity can be confirmed in vivo, for example, by administering the antibody to a nude mouse with a transplanted cancer cell line highly expressing the target protein, and determining change in the cancer cell.
  • the antibody-drug conjugate exerts an antitumor effect
  • the antibody should have the property of internalizing to migrate into cancer cells.
  • the antibody in the antibody-drug conjugate used in the present invention can be obtained by a procedure known in the art.
  • the antibody of the present invention can be obtained using a method usually carried out in the art, which involves immunizing animals with an antigenic polypeptide and collecting and purifying antibodies produced in vivo.
  • the origin of the antigen is not limited to humans, and the animals may be immunized with an antigen derived from a non-human animal such as a mouse, a rat and the like.
  • the cross-reactivity of antibodies binding to the obtained heterologous antigen with human antigens can be tested to screen for an antibody applicable to a human disease.
  • antibody-producing cells which produce antibodies against the antigen are fused with myeloma cells according to a method known in the art (e.g., Kohler and Milstein, Nature (1975) 256, p. 495-497 ; and Kennet, R. ed., Monoclonal Antibodies, p. 365-367, Plenum Press, N.Y. (1980 )) to establish hybridomas, from which monoclonal antibodies can in turn be obtained.
  • a method known in the art e.g., Kohler and Milstein, Nature (1975) 256, p. 495-497 ; and Kennet, R. ed., Monoclonal Antibodies, p. 365-367, Plenum Press, N.Y. (1980 )
  • the antigen can be obtained by genetically engineering host cells to produce a gene encoding the antigenic protein. Specifically, vectors that permit expression of the antigen gene are prepared and transferred to host cells so that the gene is expressed. The antigen thus expressed can be purified.
  • the antibody can also be obtained by a method of immunizing animals with the above-described genetically engineered antigenexpressing cells or a cell line expressing the antigen.
  • the antibody in the antibody-drug conjugate used in the present invention is preferably a recombinant antibody obtained by artificial modification for the purpose of decreasing heterologous antigenicity to humans such as a chimeric antibody or a humanized antibody, or is preferably an antibody having only the gene sequence of an antibody derived from a human, that is, a human antibody.
  • a recombinant antibody obtained by artificial modification for the purpose of decreasing heterologous antigenicity to humans
  • a chimeric antibody or a humanized antibody or is preferably an antibody having only the gene sequence of an antibody derived from a human, that is, a human antibody.
  • chimeric antibody an antibody in which antibody variable and constant regions are derived from different species, for example, a chimeric antibody in which a mouse- or rat-derived antibody variable region is connected to a human-derived antibody constant region can be exemplified ( Proc. Natl. Acad. Sci. USA, 81, 6851-6855, (1984 )).
  • an antibody obtained by integrating only the complementarity determining region (CDR) of a heterologous antibody into a human-derived antibody ( Nature (1986) 321, pp. 522-525 ), and an antibody obtained by grafting a part of the amino acid residues of the framework of a heterologous antibody as well as the CDR sequence of the heterologous antibody to a human antibody by a CDR-grafting method (International Publication No. WO 90/07861 ), and an antibody humanized using a gene conversion mutagenesis strategy ( U.S. Patent No. 5821337 ) can be exemplified.
  • CDR complementarity determining region
  • human antibody an antibody generated by using a human antibody-producing mouse having a human chromosome fragment including genes of a heavy chain and light chain of a human antibody (see Tomizuka, K. et al., Nature Genetics (1997) 16, p.133-143 ; Kuroiwa, Y. et. al., Nucl. Acids Res. (1998) 26, p.3447-3448 ; Yoshida, H. et. al., Animal Cell Technology:Basic and Applied Aspects vol.10, p.69-73 (Kitagawa, Y., Matsuda, T. and Iijima, S. eds.), Kluwer Academic Publishers, 1999 ; Tomizuka, K.
  • an antibody obtained by phage display can be exemplified.
  • an antibody obtained by phage display the antibody being selected from a human antibody library (see Wormstone, I. M. et. al, Investigative Ophthalmology & Visual Science. (2002)43 (7), p.2301-2308 ; Mé, S. et. al., Briefings in Functional Genomics and Proteomics (2002), 1(2), p.189-203 ; Siriwardena, D. et. al., Ophthalmology (2002) 109(3), p.427-431 , etc.) can be exemplified.
  • modified variants of the antibody are also included.
  • the modified variant refers to a variant obtained by subjecting the antibody according to the present invention to chemical or biological modification.
  • Examples of the chemically modified variant include variants including a linkage of a chemical moiety to an amino acid skeleton, variants including a linkage of a chemical moiety to an N-linked or O-linked carbohydrate chain, etc.
  • the biologically modified variant examples include variants obtained by post-translational modification (such as N-linked or O-linked glycosylation, N- or C-terminal processing, deamidation, isomerization of aspartic acid, or oxidation of methionine), and variants in which a methionine residue has been added to the N terminus by being expressed in a prokaryotic host cell.
  • an antibody labeled so as to enable the detection or isolation of the antibody or an antigen according to the present invention for example, an enzyme-labeled antibody, a fluorescence-labeled antibody, and an affinity-labeled antibody are also included in the meaning of the modified variant.
  • Such a modified variant of the antibody according to the present invention is useful for improving the stability and blood retention of the antibody, reducing the antigenicity thereof, detecting or isolating an antibody or an antigen, and so on.
  • antibodies subjected to such modification and functional fragments of the antibody are also included, and deletion variants in which one or two amino acids have been deleted at the carboxyl terminus of the heavy chain, variants obtained by amidation of deletion variants (for example, a heavy chain in which the carboxyl terminal proline residue has been amidated), and the like are also included.
  • the type of deletion variant having a deletion at the carboxyl terminus of the heavy chain of the antibody according to the present invention is not limited to the above variants as long as the antigen-binding affinity and the effector function are conserved.
  • the two heavy chains constituting the antibody according to the present invention may be of one type selected from the group consisting of a full-length heavy chain and the above-described deletion variant, or may be of two types in combination selected therefrom.
  • the ratio of the amount of each deletion variant can be affected by the type of cultured mammalian cells which produce the antibody according to the present invention and the culture conditions.
  • an antibody in which one amino acid residue at the carboxyl terminus has been deleted in both of the two heavy chains in the antibody according to the present invention can be preferably exemplified.
  • IgG immunoglobulin G
  • IgG1, IgG2, IgG3, IgG4 can be exemplified.
  • IgG1 or IgG2 can be exemplified.
  • antibodies in the antibody-drug conjugate used in the present invention include, but are not limited to, an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, an anti-CDH6 antibody, an anti-CD3 antibody, an anti-CD30 antibody, an anti-CD33 antibody, an anti-CD37 antibody, an anti-CD56 antibody, an anti-CD98 antibody, an anti-DR5 antibody, an anti-EGFR antibody, an anti-EPHA2 antibody, an anti-FGFR2 antibody, an anti-FGFR4 antibody, an anti-FOLR1 antibody, an anti-VEGF antibody, an anti-CD20 antibody, an anti-CD22 antibody, an anti-CD70 antibody, an anti-PSMA antibody, an anti-CEA antibody, an anti-Mesothelin antibody, an anti-A33 antibody, an anti-CanAg antibody, an anti-Cripto antibody, an anti-G250 antibody, an anti-MUC1 antibody, an anti-GPNMB antibody, an anti-
  • anti-HER2 antibody refers to an antibody which binds specifically to HER2 (Human Epidermal Growth Factor Receptor Type 2; ErbB-2), and preferably has an activity of internalization in HER2-expressing cells by binding to HER2.
  • trastuzumab U.S. Patent No. 5821337
  • pertuzumab International Publication No. WO 01/00245
  • trastuzumab can be exemplified.
  • anti-HER3 antibody refers to an antibody which binds specifically to HER3 (Human Epidermal Growth Factor Receptor Type 3; ErbB-3), and preferably has an activity of internalization in HER3-expressing cells by binding to HER3.
  • HER3 Human Epidermal Growth Factor Receptor Type 3; ErbB-3
  • anti-HER3 antibody examples include patritumab (U3-1287), U1-59 (International Publication No. WO 2007/077028 ), MM-121 (seribantumab), an anti-ERBB3 antibody described in International Publication No. WO 2008/100624 , RG-7116 (lumretuzumab), and LJM-716 (elgemtumab).
  • patritumab and U1-59 can be exemplified.
  • anti-TROP2 antibody refers to an antibody which binds specifically to TROP2 (TACSTD2: Tumor-associated calcium signal transducer 2; EGP-1), and preferably has an activity of internalization in TROP2-expressing cells by binding to TROP2.
  • anti-TROP2 antibody examples include hTINA1-H1L1 (International Publication No. WO 2015/098099 ).
  • anti-B7-H3 antibody refers to an antibody which binds specifically to B7-H3 (B cell antigen #7 homolog 3; PD-L3; CD276), and preferably has an activity of internalization in B7-H3-expressing cells by binding to B7-H3.
  • anti-B7-H3 antibody examples include M30-H1-L4 (International Publication No. WO 2014/057687 ).
  • anti-CDH6 antibody refers to an antibody which binds specifically to CDH6 (Cadherin-6), and preferably has an activity of internalization in CDH6-expressing cells by binding to CDH6.
  • anti-CDH6 antibody examples include H01L02 (International Publication No. WO 2018/212136 ).
  • a drug-linker intermediate for use in production of the antibody-drug conjugate according to the present invention is represented by the following formula.
  • the drug-linker intermediate can be expressed as the chemical name N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]glycylglycyl-L-phenylalanyl-N-[(2- ⁇ [(1S,9S)-9-ethyl-5-fluoro-9-hydroxy-4-methyl-10,13-dioxo-2,3,9,10,13,15-hexahydro-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-1-yl]amino ⁇ -2-oxoethoxy)methyl]glycinamide, and can be produced with reference to descriptions in International Publication No.
  • the antibody-drug conjugate used in the present invention can be produced by reacting the above-described drug-linker intermediate and an antibody having a thiol group (alternatively referred to as a sulfhydryl group).
  • the antibody having a sulfhydryl group can be obtained by a method well known in the art ( Hermanson, G. T, Bioconjugate Techniques, pp. 56-136, pp. 456-493, Academic Press (1996 )). For example, by using 0.3 to 3 molar equivalents of a reducing agent such as tris(2-carboxyethyl)phosphine hydrochloride (TCEP) per interchain disulfide within the antibody and reacting with the antibody in a buffer solution containing a chelating agent such as ethylenediamine tetraacetic acid (EDTA), an antibody having a sulfhydryl group with partially or completely reduced interchain disulfides within the antibody can be obtained.
  • a reducing agent such as tris(2-carboxyethyl)phosphine hydrochloride (TCEP) per interchain disulfide within the antibody
  • TCEP tris(2-carboxyethyl)phosphine hydrochloride
  • an antibody-drug conjugate in which 2 to 8 drug molecules are conjugated per antibody molecule can be produced.
  • the average number of conjugated drug molecules per antibody molecule of the antibody-drug conjugate produced can be determined, for example, by a method of calculation based on measurement of UV absorbance for the antibody-drug conjugate and the conjugation precursor thereof at two wavelengths of 280 nm and 370 nm (UV method), or a method of calculation based on quantification through HPLC measurement for fragments obtained by treating the antibody-drug conjugate with a reducing agent (HPLC method).
  • UV method UV absorbance for the antibody-drug conjugate and the conjugation precursor thereof at two wavelengths of 280 nm and 370 nm
  • HPLC method a method of calculation based on quantification through HPLC measurement for fragments obtained by treating the antibody-drug conjugate with a reducing agent
  • anti-HER2 antibody-drug conjugate refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate according to the present invention is an anti-HER2 antibody.
  • the anti-HER2 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2;
  • the average number of units of the drug-linker conjugated per antibody molecule in the anti-HER2 antibody-drug conjugate is preferably 2 to 8, more preferably 3 to 8, even more preferably 7 to 8, even more preferably 7.5 to 8, and even more preferably about 8.
  • the anti-HER2 antibody-drug conjugate used in the present invention can be produced with reference to descriptions in International Publication No. WO 2015/115091 and so on.
  • anti-HER3 antibody-drug conjugate refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate according to the present invention is an anti-HER3 antibody.
  • the anti-HER3 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 35 of SEQ ID NO: 3, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 50 to 65 of SEQ ID NO: 3, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 98 to 106 of SEQ ID NO: 3, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 24 to 39 of SEQ ID NO: 4, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 56 to 62 of SEQ ID NO: 4, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 95 to 103 of SEQ ID NO: 4;
  • the average number of units of the drug-linker conjugated per antibody molecule in the anti-HER3 antibody-drug conjugate is preferably 2 to 8, more preferably 3 to 8, even more preferably 7 to 8, even more preferably 7.5 to 8, and even more preferably about 8.
  • the anti-HER3 antibody-drug conjugate can be produced with reference to descriptions in International Publication No. WO 2015/155998 and so on.
  • anti-TROP2 antibody-drug conjugate refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
  • the anti-TROP2 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 50 to 54 of SEQ ID NO: 5, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 69 to 85 of SEQ ID NO: 5, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 118 to 129 of SEQ ID NO: 5, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 44 to 54 of SEQ ID NO: 6, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 70 to 76 of SEQ ID NO: 6, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 109 to 117 of SEQ ID NO: 6;
  • the average number of units of the drug-linker conjugated per antibody molecule in the anti-TROP2 antibody-drug conjugate used in the present invention is preferably 2 to 8, more preferably 3 to 5, even more preferably 3.5 to 4.5, and even more preferably about 4.
  • the anti-TROP2 antibody-drug conjugate can be produced with reference to descriptions in International Publication No. WO 2015/098099 , International Publication No. WO 2017/002776 , and so on.
  • anti-B7-H3 antibody-drug conjugate refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate according to the present invention is an anti-B7-H3 antibody.
  • the anti-B7-H3 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 50 to 54 of SEQ ID NO: 7, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 69 to 85 of SEQ ID NO: 7, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 118 to 130 of SEQ ID NO: 7, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 44 to 53 of SEQ ID NO: 8, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 69 to 75 of SEQ ID NO: 8, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 108 to 116 of SEQ ID NO: 8;
  • the average number of units of the drug-linker conjugated per antibody molecule in the anti-B7-H3 antibody-drug conjugate is preferably 2 to 8, more preferably 3 to 5, even more preferably 3.5 to 4.5, and even more preferably about 4.
  • the anti-B7-H3 antibody-drug conjugate used in the present invention can be produced with reference to descriptions in International Publication No. WO 2014/057687 , International Publication No. WO 2017/002776 , and so on.
  • anti-CDH6 antibody-drug conjugate refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
  • the anti-CDH6 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 45 to 54 of SEQ ID NO: 9, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 69 to 78 of SEQ ID NO: 9, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 118 to 130 of SEQ ID NO: 9, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 44 to 54 of SEQ ID NO: 10, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 70 to 76 of SEQ ID NO: 10, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 109 to 116 of SEQ ID NO: 10;
  • the average number of units of the drug-linker conjugated per antibody molecule in the anti-CDH6 antibody-drug conjugate used in the present invention is preferably 2 to 8, more preferably 3 to 8, even more preferably 7.5 to 8, and even more preferably about 8.
  • the anti-CDH6 antibody-drug conjugate used in the present invention can be produced with reference to descriptions in International Publication No. WO 2018/212136 and so on.
  • the term "kinase inhibitor” refers to an agent that inhibits a kinase involved in cancer cell growth and tumor angiogenesis.
  • the kinase inhibitor in the present invention at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2
  • the term "pharmacologically acceptable salt" of the kinase inhibitor used in the present invention may be either an acid addition salt or a base addition salt.
  • the acid addition salt can include lower alkanesulfonates such as camsylate (camphorsulfonate), mesylate (methanesulfonate), trifluoromethanesulfonate, and ethanesulfonate; arylsulfonates such as tosylate (p-toluenesulfonate) and benzenesulfonate; inorganic acid salts such as phosphate, nitrate, perchlorate, and sulfate; hydrogen halide salts such as hydrochloride, hydrobromide, hydroiodide, and hydrofluoride; organic acid salts such as acetate, malate, fumarate, succinate, citrate, tartrate, oxalate, and maleate; and amino acid salts such as ornithinate, glut
  • the base addition salt can include alkali metal salts such as sodium salt, potassium salt, and lithium salt; alkali earth metal salts such as calcium salt and magnesium salt; inorganic salts such as ammonium salt; organic amine salts such as dibenzylamine salt, morpholine salt, phenylglycine alkyl ester salt, ethylenediamine salt, N-methylglucamine salt, diethylamine salt, triethylamine salt, cyclohexylamine salt, dicyclohexylamine salt, N,N'-dibenzylethylenediamine salt, diethanolamine salt, N-benzyl-N-(2-phenylethoxy)amine salt, piperazine salt, tetramethylammonium salt, and tris(hydroxymethyl)aminomethane salt; and amino acid salts such as alginate.
  • alkali metal salts such as sodium salt, potassium salt, and lithium salt
  • alkali earth metal salts such as calcium salt and magnesium salt
  • the kinase inhibitor and pharmacologically acceptable salts thereof used in the present invention may each exist as a solvate, and solvates of them are also included in the meaning of the kinase inhibitor and pharmacologically acceptable salts thereof used in the present invention.
  • CDK4/6 inhibitor refers to an agent that inhibits cyclin dependent kinase 4 (CDK4) and inhibits cyclin dependent kinase 6 (CDK6).
  • the CDK4/6 inhibitor in the present invention may have an effect of inhibiting a kinase other than CDK4/6.
  • the CDK4/6 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include abemaciclib ( U.S. Patent No. 7855211 ), palbociclib ( U.S. Patent No. 6936612 ), ribociclib ( U.S. Patent No. 9193732 ), trilaciclib (International Publication No.
  • WO 2012/061156 G1T38 (Oncotarget 2017, 8(26): 42343-42358), PF-06873600 (International Publication No. WO 2018/033815 ), TP-1287 (International Publication No. WO 2016/187316 ), FN-1501 ( J Med Chem 2018, 61(4): 1499-1518 ), and KRX-0601 (International Publication No. WO 1989/007105 ), and pharmacologically acceptable salts thereof.
  • PF-06873600 and FN-1501 and pharmacologically acceptable salts thereof are each also known as a CDK2 inhibitor.
  • TP-1287 and pharmacologically acceptable salts thereof are each also known as a CDK1 inhibitor and a CDK2 inhibitor.
  • KRX-0601 and pharmacologically acceptable salts thereof are each also known as a PI3K inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, and a CHK1 inhibitor.
  • mTOR inhibitor refers to an agent that inhibits mTOR (Mammalian target of Rapamycin), one of the serine/threonine kinases.
  • the mTOR inhibitor in the present invention may have an effect of inhibiting a kinase other than mTOR.
  • the mTOR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include everolimus ( U.S. Patent No. 5665772 ), sirolimus ( U.S. Patent No. 5989591 ), temsirolimus ( U.S. Patent No.
  • Dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, and PQR309, and pharmacologically acceptable salts thereof are each also known as a PI3K inhibitor.
  • PI3K inhibitor refers to an agent that inhibits PI3K (Phosphoinositide 3-kinase), an enzyme that phosphorylates the hydroxyl group at position 3 of the inositol ring of inositol phospholipid.
  • the PI3K inhibitor in the present invention may have an effect of inhibiting a kinase other than PI3K.
  • the PI3K inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include taselisib (International Publication No. WO 2014/140073 ), alpelisib (International Publication No.
  • WO 2017/001645 dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, and rigosertib (International Publication No. WO 2006/010152 ), and pharmacologically acceptable salts thereof.
  • Rigosertib and pharmacologically acceptable salts thereof are each also known as a CDK1 inhibitor and a PLK1 inhibitor.
  • the pharmacologically acceptable salt of copanlisib is preferably a hydrochloride (copanlisib hydrochloride).
  • the pharmacologically acceptable salt of rigosertib is preferably a sodium salt (rigosertib sodium).
  • the term "AKT inhibitor” refers to an agent that inhibits AKT (also called protein kinase B), a serine/threonine kinase.
  • AKT inhibitor in the present invention may have an effect of inhibiting a kinase other than AKT.
  • the AKT inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ipatasertib (International Publication No. WO 2008/006040 ), uprosertib (International Publication No.
  • the pharmacologically acceptable salt of ipatasertib is preferably a hydrochloride (ipatasertib hydrochloride).
  • the term "ERK inhibitor” refers to an agent that inhibits extracellular signal-regulated kinase (ERK).
  • the ERK inhibitor in the present invention may have an effect of inhibiting a kinase other than ERK.
  • the ERK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ONC201 ( Oncotarget 2014, 5(24): 12728-37 ), BVD-523 ( Mol Cancer Ther 2017, 16(11): 2351-2363 ), CC-90003 (International Publication No. WO 2014/124230 ), GDC-0994 ( J Med Chem 2016, 59(12): 5650-5660 ), LY3214996 (International Publication No. WO 2016/106029 ), and MK-8353 ( ACS Med Chem Lett 2018, 9(7): 761-767 ), and pharmacologically acceptable salts thereof.
  • the term "MEK inhibitor” refers to an agent that inhibits mitogen-activated extracellular signal-regulated kinase (MEK).
  • MEK mitogen-activated extracellular signal-regulated kinase
  • the MEK inhibitor in the present invention may have an effect of inhibiting a kinase other than MEK.
  • the MEK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include trametinib ( U.S. Patent No. 7378423 ), binimetinib ( U.S. Patent No. 7777050 ), selumetinib (International Publication No. WO 2005/023251 ), refametinib (International Publication No. WO 2007/014011 ), pimasertib (International Publication No.
  • Trametinib is preferably a dimethyl sulfoxide adduct (trametinib dimethyl sulfoxide).
  • the term "RAF inhibitor” refers to an agent that inhibits the kinase activity of RAF (preferably, BRAF).
  • the RAF inhibitor in the present invention may have an effect of inhibiting a kinase other than RAF.
  • the RAF inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib ( U.S. Patent No. 7351834 ), sorafenib ( U.S. Patent No. 7235576 ), vemurafenib ( U.S. Patent No. 7504509 ), dabrafenib ( U.S. Patent No. 7994185 ), encorafenib ( U.S.
  • Patent No. 8501758 discloses RAF265 (ACS Med Chem Lett 2015, 6(9): 961-5 ), GDC-5573 (International Publication No. WO 2013/100632 ), LY3009120 ( J Med Chem 2015, 58(10): 4165-4179 ), and RO5126766, and pharmacologically acceptable salts thereof.
  • Regorafenib and pharmacologically acceptable salts thereof are each also known as a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, and an FGFR inhibitor.
  • Sorafenib and pharmacologically acceptable salts thereof are each also known as a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, and an FLT3 inhibitor.
  • Regorafenib is preferably a hydrate (regorafenib hydrate).
  • the pharmacologically acceptable salt of sorafenib is preferably a tosylate (sorafenib tosylate).
  • the pharmacologically acceptable salt of dabrafenib is preferably a mesylate (dabrafenib mesylate).
  • CDK1 inhibitor refers to an agent that inhibits cyclin dependent kinase 1 (CDK1).
  • CDK1 inhibitor in the present invention may have an effect of inhibiting a kinase other than CDK1.
  • the CDK1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include dinaciclib (International Publication No. WO 2005/077954 ), milciclib ( J Med Chem 2009, 52(16): 5152-63 ), seliciclib (International Publication No. WO 1997/020842 ), alvocidib (International Publication No. WO 2001/053293 ), roniciclib (International Publication No.
  • CDK2 inhibitor refers to an agent that inhibits cyclin dependent kinase 2 (CDK2).
  • CDK2 inhibitor in the present invention may have an effect of inhibiting a kinase other than CDK2.
  • the CDK2 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include CYC-065, PF-06873600, FN-1501, KRX-0601, dinaciclib, seliciclib, alvocidib, voruciclib, AT7519, PHA-793887, and roniciclib, and pharmacologically acceptable salts thereof.
  • CHK1 inhibitor refers to an agent that inhibits checkpoint kinase 1 (CHK1).
  • CHK1 inhibitor in the present invention may have an effect of inhibiting a kinase other than CHK1.
  • the CHK1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include MK-8776 ( Bioorg Med Chem Lett 2011, 21(1): 471-4 ), LY2606368 ( Mol Cancer Ther 2015, 14(9): 2004-13 ), LY2603618 ( Invest New Drugs 2013, 31(1): 136-44 ), CBP501 ( Mol Cancer Ther 2007, 6(1): 147-53 ), GDC-0425 ( Org Process Res Dev 2015, 19(6): 661-672 ), CCT245737 ( Oncotarget 2016, 7(3): 2329-42 ), and KRX-0601, and pharmacologically acceptable salts thereof.
  • the term "WEE1 inhibitor” refers to an agent that inhibits Wee1-like protein kinase.
  • the WEE1 inhibitor in the present invention may have an effect of inhibiting a kinase other than WEE1.
  • the WEE1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include milciclib and MK-1775 ( Mol Cancer Ther 2009, 8(11): 2992-3000 ), and pharmacologically acceptable salts thereof.
  • the term "PLK1 inhibitor” refers to an agent that inhibits Polo-like kinase 1 (PLK1).
  • the PLK1 inhibitor in the present invention may have an effect of inhibiting a kinase other than PLK1.
  • the PLK1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include rigosertib and volasertib (International Publication No. WO 2012/049153 ), and pharmacologically acceptable salts thereof.
  • the term "Aurora kinase inhibitor” refers to an agent that inhibits Aurora kinase.
  • the Aurora kinase inhibitor in the present invention may have an effect of inhibiting a kinase other than Aurora kinase.
  • the Aurora kinase inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include alisertib (International Publication No. WO 2005/111039 ), ilorasertib (International Publication No. WO 2010/065825 ), ENMD-2076 ( Clin Cancer Res 2011, 17(4): 849-60 ), AMG900 ( Cancer Res 2010, 70(23): 9846-54 ), and roniciclib, and pharmacologically acceptable salts thereof.
  • Ilorasertib and pharmacologically acceptable salts thereof are each also known as a VEGFR inhibitor, a KIT inhibitor, a PDGFR inhibitor, an FLT3 inhibitor, and a CSF-1R inhibitor.
  • ENMD-2076 is also known as a VEGFR inhibitor and an FLT3 inhibitor.
  • Bcr-Abl inhibitor refers to an agent that inhibits the tyrosine kinase Bcr-Abl.
  • the Bcr-Abl inhibitor in the present invention may have an effect of inhibiting a kinase other than Bcr-Abl.
  • the Bcr-Abl inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include imatinib ( U.S. Patent No. 6894051 ), dasatinib ( U.S. Patent No. 6596746 ), bosutinib ( U.S. Patent No. 6002008 ), nilotinib ( U.S. Patent No. 7169791 ), and ponatinib ( U.S. Patent No. 8114874 ), and pharmacologically acceptable salts thereof.
  • Imatinib and nilotinib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor and a PDGFR inhibitor.
  • Dasatinib and pharmacologically acceptable salts thereof are each also known as an Src inhibitor, a KIT inhibitor, and a PDGFR inhibitor.
  • Bosutinib and pharmacologically acceptable salts thereof are each also known as an Src inhibitor.
  • Ponatinib and pharmacologically acceptable salts thereof are each also known as an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, and an FLT3 inhibitor.
  • the pharmacologically acceptable salt of imatinib is preferably mesylate (imatinib mesylate).
  • Dasatinib is preferably a hydrate (dasatinib hydrate).
  • Bosutinib is preferably a hydrate (bosutinib hydrate).
  • the pharmacologically acceptable salt of nilotinib is preferably a hydrochloride hydrate (nilotinib hydrochloride hydrate).
  • the pharmacologically acceptable salt of ponatinib is preferably a hydrochloride (ponatinib hydrochloride).
  • the term "Src inhibitor” refers to an agent that inhibits the protooncogene tyrosine protein kinase Src.
  • the Src inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include dasatinib and bosutinib, and pharmacologically acceptable salts thereof.
  • EPH inhibitor refers to an agent that inhibits tyrosine kinase of an erythropoietin-producing hepatocellular receptor (EPH).
  • EPH inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include dasatinib and ponatinib, and pharmacologically acceptable salts thereof.
  • the term "VEGFR inhibitor” refers to an agent that inhibits tyrosine kinase of a vascular endothelial growth factor receptor (VEGFR).
  • VEGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than VEGFR tyrosine kinase.
  • the VEGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, vandetanib ( U.S. Patent No. 7173038 ), sunitinib ( U.S. Patent No. 6573293 ), axitinib ( U.S. Patent No.
  • pazopanib U.S. Patent No. 7105530
  • lenvatinib U.S. Patent No. 7253286
  • nintedanib hereinafter, also referred to as "BIBF1120"
  • U.S. Patent No. 6762180 U.S. Patent No. 6762180
  • cabozantinib U.S. Patent No. 7579473
  • tivozanib International Publication No. WO 2002/088110
  • brivanib International Publication No. WO 2004/009784
  • linifanib International Publication No. WO 2014/022975
  • lucitanib International Publication No. WO 2008/112408
  • ilorasertib and ENMD-2076, and pharmacologically acceptable salts thereof.
  • Vandetanib and pharmacologically acceptable salts thereof are each also known as an RET inhibitor.
  • Sunitinib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FLT3 inhibitor, and a CSF-1R inhibitor.
  • Axitinib and pharmacologically acceptable salts thereof are each also known as a PDGFR inhibitor, an FGFR inhibitor, and a CSF-1R inhibitor.
  • Pazopanib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor, a PDGFR inhibitor, and an FGFR inhibitor.
  • Lenvatinib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, and an FGFR inhibitor.
  • Nintedanib and pharmacologically acceptable salts thereof are each also known as a PDGFR inhibitor and an FGFR inhibitor.
  • the pharmacologically acceptable salt of sunitinib is preferably a malate (sunitinib malate).
  • the pharmacologically acceptable salt of pazopanib is preferably a hydrochloride (pazopanib hydrochloride).
  • the pharmacologically acceptable salt of lenvatinib is preferably a mesylate (lenvatinib mesylate).
  • the pharmacologically acceptable salt of nintedanib is preferably an ethanesulfonate (nintedanib ethanesulfonate).
  • the pharmacologically acceptable salt of cabozantinib is preferably a malate (cabozantinib malate).
  • the pharmacologically acceptable salt of tivozanib is preferably a hydrochloride hydrate (tivozanib hydrochloride hydrate).
  • KIT inhibitor refers to an agent that inhibits tyrosine kinase of the cytokine receptor KIT (c-kit, or also called CD117).
  • the KIT inhibitor in the present invention may have an effect of inhibiting a kinase other than KIT tyrosine kinase.
  • the KIT inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, and dasatinib, and pharmacologically acceptable salts thereof.
  • the term "RET inhibitor” refers to an agent that inhibits RET (Rearranged during transfection) tyrosine kinase.
  • the RET inhibitor in the present invention may have an effect of inhibiting a kinase other than RET tyrosine kinase.
  • the RET inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, vandetanib, lenvatinib, and sunitinib, and pharmacologically acceptable salts thereof.
  • the term "PDGFR inhibitor” refers to an agent that inhibits tyrosine kinase of a platelet-derived growth factor receptor (PDGFR).
  • PDGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than PDGFR tyrosine kinase.
  • the PDGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, and dasatinib, and pharmacologically acceptable salts thereof.
  • FGFR inhibitor refers to an agent that inhibits tyrosine kinase of a fibroblast growth factor receptor (FGFR).
  • FGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than FGFR tyrosine kinase.
  • the FGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, and pazopanib, and pharmacologically acceptable salts thereof.
  • the term "BTK inhibitor” refers to an agent that inhibits Bruton's tyrosine kinase (BTK).
  • the BTK inhibitor in the present invention may have an effect of inhibiting a kinase other than BTK.
  • the BTK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ibrutinib ( U.S. Patent No. 7514444 ), acalabrutinib ( U.S. Patent No. 9290504 ), and tirabrutinib (International Publication No. WO 2011/152351 ), and pharmacologically acceptable salts thereof.
  • the term "FLT3 inhibitor” refers to an agent that inhibits FMS-like tyrosine kinase 3 (FLT3).
  • the FLT3 inhibitor in the present invention may have an effect of inhibiting a kinase other than FLT3.
  • the FLT3 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include gilteritinib ( U.S. Patent No. 7514444 ), quizartinib ( J Med Chem 2009, 52(23): 7808-7816 ), midostaurin (International Publication No. WO 2003/037347 ), sorafenib, ilorasertib, ENMD-2076, and sunitinib, and pharmacologically acceptable salts thereof.
  • the pharmacologically acceptable salt of gilteritinib is preferably a fumarate (gilteritinib fumarate).
  • the term "ALK inhibitor” refers to an agent that inhibits anaplastic lymphoma kinase (ALK).
  • the ALK inhibitor in the present invention may have an effect of inhibiting a kinase other than ALK.
  • the ALK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include brigatinib ( U.S. Patent No. 9012462 ), crizotinib ( U.S. Patent No. 7858643 ), ceritinib ( U.S. Patent No. 7153964 ), alectinib ( U.S. Patent No. 9126931 ), and lorlatinib ( U.S. Patent No. 8680111 ), and pharmacologically acceptable salts thereof.
  • the pharmacologically acceptable salt of alectinib is preferably a hydrochloride (alectinib hydrochloride).
  • the term "JAK inhibitor” refers to an agent that inhibits Janus kinase (JAK).
  • the JAK inhibitor in the present invention may have an effect of inhibiting a kinase other than JAK.
  • the JAK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ruxolitinib ( U.S. Patent No. 7598257 ), tofacitinib ( U.S. Patent No. 7265221 ), baricitinib ( U.S. Patent No. 8158616 ), and pacritinib (International Publication No. WO 2007/058627 ), and pharmacologically acceptable salts thereof.
  • the pharmacologically acceptable salt of ruxolitinib is preferably a phosphate (ruxolitinib phosphate).
  • the pharmacologically acceptable salt of tofacitinib is preferably a citrate (tofacitinib citrate).
  • MET inhibitor refers to an agent that inhibits MET (also referred to as "c-Met"), a receptor-type tyrosine kinase whose ligand is a hepatocyte growth factor (HGF).
  • HGF hepatocyte growth factor
  • the MET inhibitor in the present invention may have an effect of inhibiting a kinase other than MET.
  • the MET inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include capmatinib (International Publication No. WO 2008/064157 ) and tepotinib (International Publication No. WO 2009/006959 ), and pharmacologically acceptable salts thereof.
  • CSF-1R inhibitor refers to an agent that inhibits tyrosine kinase of a colony-stimulating factor-1 receptor (CSF-1R).
  • CSF-1R inhibitor in the present invention may have an effect of inhibiting a kinase other than tyrosine kinase of CSF-1R.
  • the CSF-1R inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include pexidartinib ( Clin Cancer Res 2014, 20(12): 3146-58 ), BLZ-945 (International Publication No. WO 2007/121484 ), JNJ-40346527 (International Publication No.
  • JNJ-28312141 J Med Chem 2011, 54(22): 7860-7883
  • ilorasertib imatinib, sunitinib, and axitinib, and pharmacologically acceptable salts thereof.
  • the pharmacologically acceptable salt of pexidartinib is preferably a hydrochloride (pexidartinib hydrochloride).
  • NTRK inhibitor refers to an agent that inhibits neurotrophic tropomyosin receptor kinase (TRK).
  • TRK neurotrophic tropomyosin receptor kinase
  • the NTRK inhibitor in the present invention may have an effect of inhibiting a kinase other than TRK.
  • the NTRK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include entrectinib (International Publication No. WO 2009/013126 ), and GR-389988 (International Publication No. WO 2015/089139 ), and pharmacologically acceptable salts thereof.
  • the term "EGFR inhibitor” refers to an agent that inhibits tyrosine kinase of an epidermal growth factor receptor (EGFR).
  • the EGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than tyrosine kinase of EGFR.
  • the EGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include gefitinib (International Publication No. WO 1996/033980 ), erlotinib (International Publication No. WO 1996/030347 ), afatinib (International Publication No. WO 2002/050043 ), osimertinib (International Publication No.
  • WO 2013/014448 dacomitinib ( U.S. Patent No. 7772243 ), lapatinib ( U.S. Patent No. 713485 ), neratinib ( J Med Chem 2005, 48(4): 1107-1131 ), pyrotinib (International Publication No. WO 2011/029265 ), and poziotinib (International Publication No. WO 2014/116070 ), and pharmacologically acceptable salts thereof.
  • the term "HER2 inhibitor” refers to an agent that inhibits tyrosine kinase of HER2 (human epidermal growth factor receptor type 2; ErbB-2).
  • the HER2 inhibitor in the present invention may have an effect of inhibiting a kinase other than tyrosine kinase of HER2.
  • the HER2 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include tucatinib (International Publication No. WO 2013/056183 ), neratinib, mubritinib (International Publication No. WO 2001/077107 ), lapatinib, pyrotinib, and poziotinib, and pharmacologically acceptable salts thereof.
  • the pharmaceutical composition and method of treatment of the present invention may be characterized in that the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times, or characterized in that the antibody-drug conjugate and the kinase inhibitor are contained as active components in a single formulation and administered.
  • two or more of the kinase inhibitors used in the present invention can be administered in combination.
  • the pharmaceutical composition and method of treatment of the present invention can be used for treating cancer, and can be preferably used for treating at least one cancer selected from the group consisting of breast cancer (including triple-negative breast cancer and luminal breast cancer), gastric cancer (also called gastric adenocarcinoma), colorectal cancer (also called colon and rectal cancer, and including colon cancer and rectal cancer), lung cancer (including small cell lung cancer and non-small cell lung cancer), esophageal cancer, head-and-neck cancer (including salivary gland cancer and pharyngeal cancer), gastroesophageal junction adenocarcinoma, biliary tract cancer (including bile duct cancer), Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney
  • the kind of antibody preferably used in the antibody-drug conjugate can be determined by examining the type of cancer and tumor markers. For example, if HER2 expression is found in the cancer, an anti-HER2 antibody-drug conjugate can be preferably used; if HER3 expression is found in the cancer, an anti-HER3 antibody-drug conjugate can be preferably used; if TROP2 expression is found in the cancer, an anti-TROP2 antibody-drug conjugate can be preferably used; if B7-H3 expression is found in the cancer, an anti-B7-H3 antibody-drug conjugate can be preferably used; and if CDH6 expression is found in the cancer, an anti-CDH6 antibody-drug conjugate can be preferably used.
  • the presence or absence of HER2, HER3, TROP2, B7-H3, and CDH6, and other tumor markers can be checked by, for example, collecting tumor tissue from a cancer patient, and subjecting the formalin-fixed paraffin-embedded specimen (FFPE) to an examination at a gene product (protein) level, such as an immunohistochemistry (IHC) method, a flow cytometry, a western blot method, or an examination at a gene transcription level such as an in situ hybridization method (ISH), a quantitative PCR method (q-PCR), or a microarray analysis; alternatively, it can also be checked by collecting cell-free blood circulating tumor DNA (ctDNA) from a cancer patient and subjecting it to an examination which uses a method such as next generation sequencing (NGS).
  • FFPE formalin-fixed paraffin-embedded specimen
  • IHC immunohistochemistry
  • ISH in situ hybridization method
  • q-PCR quantitative PCR method
  • NGS next generation sequencing
  • the pharmaceutical composition and method of treatment of the present invention can be preferably used for a mammal, and can be more preferably used for a human.
  • the antitumor effect of the pharmaceutical composition and method of treatment of the present invention can be confirmed by, for example, generating a model in which cancer cells are transplanted to a test animal, and measuring reduction in tumor volume or lifeprolonging effects due to applying the pharmaceutical composition and method of treatment of the present invention. Furthermore, comparison with the antitumor effect of single administrations of each of the antibody-drug conjugate and the kinase inhibitor used in the present invention can provide confirmation of the combined effect of the antibody-drug conjugate and the kinase inhibitor used in the present invention.
  • the antitumor effect of the pharmaceutical composition and method of treatment of the present invention can be confirmed, in a clinical study, with the Response Evaluation Criteria in Solid Tumors (RECIST) evaluation method, WHO's evaluation method, Macdonald's evaluation method, measurement of body weight, and other methods; and can be determined by indicators such as Complete response (CR), Partial response (PR), Progressive disease (PD), Objective response rate (ORR), Duration of response (DoR), Progression-free survival (PFS), and Overall survival (OS).
  • RECIST Response Evaluation Criteria in Solid Tumors
  • the foregoing methods can provide confirmation of superiority in terms of the antitumor effect of the pharmaceutical composition and method of treatment of the present invention compared to existing pharmaceutical compositions and methods of treatment for cancer therapy.
  • the pharmaceutical composition and method of treatment of the present invention can retard growth of cancer cells, suppress their proliferation, and further can kill cancer cells. These effects can allow cancer patients to be free from symptoms caused by cancer or can achieve an improvement in the QOL of cancer patients and attain a therapeutic effect by sustaining the lives of the cancer patients. Even if the pharmaceutical composition and method of treatment of the present invention do not accomplish the killing of cancer cells, they can achieve higher QOL of cancer patients while achieving longer-term survival, by inhibiting or controlling the growth of cancer cells.
  • the pharmaceutical composition of the present invention can be expected to exert a therapeutic effect by application as systemic therapy to patients, and additionally, by local application to cancer tissues.
  • the pharmaceutical composition of the present invention may be administered as a pharmaceutical composition containing at least one pharmaceutically suitable ingredient.
  • the pharmaceutically suitable ingredient can be suitably selected and applied from formulation additives or the like that are generally used in the art, in view of the dosage, administration concentration, or the like of the antibody-drug conjugate and the kinase inhibitor used in the present invention.
  • the antibody-drug conjugate used in the present invention may be administered as a pharmaceutical composition containing a buffer such as histidine buffer, an excipient such as sucrose and trehalose, and a surfactant such as Polysorbates 80 and 20.
  • the pharmaceutical composition containing the antibody-drug conjugate used in the present invention can be preferably used as an injection, can be more preferably used as an aqueous injection or a lyophilized injection, and can be even more preferably used as a lyophilized injection.
  • the aqueous injection can be preferably diluted with a suitable diluent and then given as an intravenous infusion.
  • a suitable diluent a dextrose solution, physiological saline, and the like, can be exemplified, and a dextrose solution can be preferably exemplified, and a 5% dextrose solution can be more preferably exemplified.
  • the pharmaceutical composition containing the antibody-drug conjugate used in the present invention is a lyophilized injection
  • it can be preferably dissolved in water for injection, subsequently a required amount can be diluted with a suitable diluent and then given as an intravenous infusion.
  • a suitable diluent a dextrose solution, physiological saline, and the like, can be exemplified, and a dextrose solution can be preferably exemplified, and a 5% dextrose solution can be more preferably exemplified.
  • Examples of the administration route which may be used to administer the pharmaceutical composition of present invention include intravenous, intradermal, subcutaneous, intramuscular, and intraperitoneal routes; and preferably include an intravenous route.
  • the antibody-drug conjugate used in the present invention can be administered to a human once at intervals of 1 to 180 days, and can be preferably administered once a week, once every 2 weeks, once every 3 weeks, or once every 4 weeks, and can be even more preferably administered once every 3 weeks. Also, the antibody-drug conjugate used in the present invention can be administered at a dose of about 0.001 to 100 mg/kg, and can be preferably administered at a dose of 0.8 to 12.4 mg/kg.
  • the antibody-drug conjugate used in the present invention is an anti-HER2 antibody-drug conjugate
  • it can be preferably administered once every 3 weeks at a dose of 0.8 mg/kg, 1.6 mg/kg, 3.2 mg/kg, 5.4 mg/kg, 6.4 mg/kg, 7.4 mg/kg, or 8 mg/kg.
  • the antibody-drug conjugate used in the present invention is an anti-HER3 antibody-drug conjugate
  • it can be preferably administered once every 3 weeks at a dose of 1.6 mg/kg, 3.2 mg/kg, 4.8 mg/kg, 6.4 mg/kg, 8.0 mg/kg, 9.6 mg/kg, or 12.8 mg/kg.
  • the antibody-drug conjugate used in the present invention is an anti-TROP2 antibody-drug conjugate
  • it can be preferably administered once every 3 weeks at a dose of 0.27 mg/kg, 0.5 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 4.0 mg/kg, 6.0 mg/kg, or 8.0 mg/kg.
  • the kinase inhibitor according to the present invention can be orally administered to a human once or twice at intervals of 1 to 7 days, and can be preferably orally administered once a day or twice per day.
  • the kinase inhibitor used in the present invention can be orally administered at a dose of 0.1 mg to 3000 mg, and can be preferably administered at a dose of 2.5 mg to 600 mg.
  • the kinase inhibitor used in the present invention can be administered to a human as an intravenous drip once at intervals of 1 to 180 days, and can be preferably administered as an intravenous drip once a week, once every 2 weeks, once every 3 weeks, or once every 4 weeks.
  • the kinase inhibitor used in the present invention can be administered as an intravenous drip at a dose of 0.1 mg to 3000 mg, and can be preferably administered as an intravenous drip at a dose of 10 mg to 100 mg.
  • the kinase inhibitor used in the present invention is abemaciclib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 50 mg, 100 mg, 150 mg, or 200 mg.
  • the kinase inhibitor used in the present invention is palbociclib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 75 mg, 100 mg, or 125 mg.
  • the kinase inhibitor used in the present invention is ribociclib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 200 mg, 400 mg, or 600 mg.
  • the kinase inhibitor used in the present invention is everolimus or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 2.5 mg, 5 mg, 7.5 mg, 10 mg, 15 mg, or 20 mg.
  • the kinase inhibitor used in the present invention is sirolimus or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 0.5 mg, 1 mg, 1.5 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 8 mg, 10 mg, 12 mg, or 15 mg.
  • the kinase inhibitor used in the present invention is temsirolimus or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably administered as an intravenous drip once a week at a dose of 25 mg.
  • the kinase inhibitor used in the present invention is copanlisib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably administered as an intravenous drip once with intervals of 1 to 3 weeks at a dose of 60 mg.
  • the kinase inhibitor used in the present invention is idelalisib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 100 mg or 150 mg.
  • the kinase inhibitor used in the present invention is duvelisib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 15 mg or 25 mg.
  • the kinase inhibitor used in the present invention is trametinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 0.5 mg, 1 mg, 1.5 mg, or 2 mg.
  • the kinase inhibitor used in the present invention is binimetinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 15 mg, 30 mg, or 45 mg.
  • the kinase inhibitor used in the present invention is cobimetinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 40 mg, or 60 mg.
  • the kinase inhibitor used in the present invention is regorafenib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg, 80 mg, 120 mg, or 160 mg.
  • the kinase inhibitor used in the present invention is sorafenib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 200 mg or 400 mg.
  • the kinase inhibitor used in the present invention is vemurafenib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 240 mg, 480 mg, 720 mg, or 960 mg.
  • the kinase inhibitor used in the present invention is dabrafenib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 50 mg, 75 mg, 100 mg, 125 mg, or 150 mg.
  • the kinase inhibitor used in the present invention is encorafenib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 50 mg, 75 mg, 100 mg, 125 mg, 150 mg, 175 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, or 450 mg.
  • the kinase inhibitor used in the present invention is imatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg, or twice per day at a dose of 400 mg.
  • the kinase inhibitor used in the present invention is dasatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 50 mg, 70 mg, 80 mg, 100 mg, or 140 mg.
  • the kinase inhibitor used in the present invention is bosutinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg.
  • the kinase inhibitor used in the present invention is nilotinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 150 mg, 200 mg, 300 mg, or 400 mg.
  • the kinase inhibitor used in the present invention is ponatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 15 mg, 30 mg, or 45 mg.
  • the kinase inhibitor used in the present invention is vandetanib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 100 mg, 200 mg, or 300 mg.
  • the kinase inhibitor used in the present invention is axitinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 1 mg, 2 mg, 3 mg, 4 mg, or 5 mg.
  • the kinase inhibitor used in the present invention is pazopanib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 200 mg, 400 mg, 600 mg, or 800 mg.
  • the kinase inhibitor used in the present invention is lenvatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 4 mg, 8 mg, 10 mg, 12 mg, 14 mg, 16 mg, 18 mg, 20 mg, or 24 mg.
  • the kinase inhibitor used in the present invention is nintedanib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 100 mg or 150 mg.
  • the kinase inhibitor used in the present invention is cabozantinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 40 mg, or 60 mg.
  • the kinase inhibitor used in the present invention is ibrutinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 70 mg, 140 mg, 210 mg, 280 mg, 350 mg, 420 mg, 490 mg, or 560 mg.
  • the kinase inhibitor used in the present invention is acalabrutinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once per 12 hours at a dose of 100 mg.
  • the kinase inhibitor used in the present invention is gilteritinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg, 80 mg, or 120 mg.
  • the kinase inhibitor used in the present invention is midostaurin or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 25 mg, 50 mg, 75 mg, or 100 mg.
  • the kinase inhibitor used in the present invention is brigatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 30 mg, 60 mg, 90 mg, 120 mg, 150 mg, or 180 mg.
  • the kinase inhibitor used in the present invention is crizotinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 200 mg or 250 mg.
  • the kinase inhibitor used in the present invention is ceritinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 150 mg, 300 mg, 450 mg, 600 mg, or 750 mg.
  • the kinase inhibitor used in the present invention is alectinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 150 mg, 300 mg, 450 mg, or 600 mg.
  • the kinase inhibitor used in the present invention is lorlatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 25 mg, 50 mg, 75 mg, or 100 mg.
  • the kinase inhibitor used in the present invention is ruxolitinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 5 mg, 10 mg, 15 mg, 20 mg, or 25 mg.
  • the kinase inhibitor used in the present invention is tofacitinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered twice per day at a dose of 5 mg or 10 mg per administration, and in using as a sustained-release formulation preferably administered once a day at a dose of 11 mg.
  • the kinase inhibitor used in the present invention is baricitinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 2 mg.
  • the kinase inhibitor used in the present invention is gefitinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 250 mg.
  • the kinase inhibitor used in the present invention is erlotinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 25 mg, 50 mg, 75 mg, 100 mg, 125 mg, or 150 mg.
  • the kinase inhibitor used in the present invention is afatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 30 mg, or 40 mg.
  • the kinase inhibitor used in the present invention is osimertinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg or 80 mg.
  • the kinase inhibitor used in the present invention is dacomitinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 15 mg, 30 mg, or 45 mg.
  • the kinase inhibitor used in the present invention is lapatinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 250 mg, 500 mg, 750 mg, 1000 mg, 1250 mg, or 1500 mg.
  • the kinase inhibitor used in the present invention is neratinib or a pharmacologically acceptable salt thereof
  • the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg, 80 mg, 120 mg, 160 mg, 200 mg, or 240 mg.
  • the pharmaceutical composition and method of treatment of the present invention may further contain a cancer therapeutic agent other than the antibody-drug conjugate and the kinase inhibitor according to the present invention.
  • the pharmaceutical composition and method of treatment of the present invention can also be administered in combination with another cancer therapeutic agent, thereby enhancing the antitumor effect.
  • Other cancer therapeutic agents to be used for such purpose may be administered to a subject simultaneously with, separately from, or subsequently with the pharmaceutical composition of the present invention, or may be administered while varying the dosage interval for each.
  • Such cancer therapeutic agents are not limited as long as they are agents having antitumor activity, and can be exemplified by at least one selected from the group consisting of irinotecan (CPT-11), cisplatin, carboplatin, oxaliplatin, fluorouracil (5-FU), gemcitabine, capecitabine, paclitaxel, docetaxel, doxorubicin, epirubicin, cyclophosphamide, mitomycin C, a tegafur-gimeracil-oteracil combination drug, cetuximab, panitumumab, bevacizumab, ramucirumab, a trifluridinetipiracil combination drug, methotrexate, pemetrexed, tamoxifen, toremifene, fulvestrant, leuprorelin, goserelin, letrozole, anastrozole, a progesterone formulation, trastuzumab, and pertu
  • the pharmaceutical composition and method of treatment of the present invention can also be used in combination with radiotherapy.
  • a cancer patient may receive radiotherapy before and/or after or simultaneously with receiving therapy with the pharmaceutical composition of the present invention.
  • the pharmaceutical composition and method of treatment of the present invention can also be used as adjuvant chemotherapy in combination with a surgical procedure.
  • the pharmaceutical composition of the present invention may be administered for the purpose of diminishing the size of a tumor before surgical procedure (referred to as pre-operative adjuvant chemotherapy or neoadjuvant therapy), or may be administered after a surgical procedure for the purpose of preventing the recurrence of a tumor (referred to as post-operative adjuvant chemotherapy or adjuvant therapy).
  • an antibody-drug conjugate in which a drug-linker represented by the following formula: wherein A represents a connecting position to an antibody, is conjugated to the anti-HER2 antibody via a thioether bond (hereinafter, referred to as the "antibody-drug conjugate (1)") was produced.
  • the DAR of the antibody-drug conjugate (1) is 7.7 or 7.8.
  • mice 5 to 6-week-old BALB/c nude mice (Charles River Laboratories Japan, Inc.) were subjected to experiment.
  • the antibody-drug conjugate (1) was diluted with ABS buffer (10 mM acetate buffer solution (pH 5.5), 5% sorbitol), and intravenously administered to the tail vein at a liquid volume of 10 mL/kg.
  • Everolimus was suspended in 30% propylene glycol and 5% Tween 80, and orally administered at a liquid volume of 10 mL/kg.
  • Taselisib was suspended in 0.5% methyl cellulose and 0.2% Tween 80, and orally administered at a liquid volume of 10 mL/kg.
  • Abemaciclib was suspended in 1% hydroxyethyl cellulose and 0.1% antifoam/25 mM phosphate buffer, pH2, and orally administered at a liquid volume of 10 mL/kg.
  • MDA-MB-453 cells a human breast cancer cell line, purchased from ATCC (American Type Culture Collection) were suspended in Matrigel basement membrane matrix, and 1 ⁇ 10 7 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0).
  • the antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 0.5 mg/kg on Day 0.
  • Everolimus was administered once a day, five times a week, at a dose of 5 mg/kg for 3 weeks.
  • Taselisib was administered once a day, five times a week, at a dose of 5 mg/kg for 3 weeks.
  • Abemaciclib was administered once a day, five times a week, at a dose of 30 mg/kg for 2 weeks.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (1) and each kinase inhibitor, and a solvent administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (1) and everolimus are shown in Figure 11 .
  • the tumor growth inhibition (TGI) of single administration of everolimus on the day of effect determination was 64%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 75%.
  • the tumor growth inhibition (TGI: 89%) was higher than those of single administration of them, demonstrating strong effect of their combined use.
  • the abscissa depicts days from the initiation of administration, and the ordinate depicts tumor volumes. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group. In evaluation examples below for antitumor tests, the tests were conducted with the procedure used in this evaluation example, unless otherwise stated.
  • results of combined use of the antibody-drug conjugate (1) and taselisib are shown in Figure 12 .
  • the TGI of single administration of taselisib was 67%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 75%.
  • the tumor growth inhibition (TGI: 92%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • results of combined use of the antibody-drug conjugate (1) and abemaciclib are shown in Figure 13 .
  • the TGI of single administration of abemaciclib was 68%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 75%.
  • the tumor growth inhibition (TGI: 87%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • JIMT-1 cells a human breast cancer cell line, purchased from DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH) were suspended in physiological saline, and 5 ⁇ 10 6 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 15 days after the transplantation (Day 0).
  • the antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 10 mg/kg on Day 0.
  • Everolimus was administered once a day, five times a week and seven times in total, at a dose of 5 mg/kg.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (1) and everolimus, and a solvent administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (1) and everolimus are shown in Figure 14 .
  • the TGI of single administration of everolimus was 52%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 53%.
  • the tumor growth inhibition (TGI: 93%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • COLO201 cells a human colorectal cancer cell line, purchased from ATCC were suspended in 50% Matrigel solution, and 5 ⁇ 10 6 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0).
  • the antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 3 mg/kg on Day 0.
  • Regorafenib was dissolved in Cremophor EL/95% ethanol and then diluted with distilled water, and orally administered at a liquid volume of 10 mL/kg. Administration was performed once a day, five times a week, at a dose of 10 mg/kg for three weeks.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (1) and regorafenib, and a solvent administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (1) and regorafenib are shown in Figure 15 .
  • the TGI of single administration of regorafenib was 81%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 67%.
  • the tumor growth inhibition (TGI: 99%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • KPL-4 cells a human breast cancer cell line, obtained from Dr. Junichi Kurebayashi, Kawasaki Medical School ( British Journal of Cancer. (1999)79 (5/6). 707-717 ) were suspended in physiological saline, and 1.5 ⁇ 10 7 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 17 days after the transplantation (Day 0).
  • the antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 7.5 mg/kg on Day 0.
  • Tucatinib was suspended in 0.5% methyl cellulose, and orally administered at a liquid volume of 10 mL/kg. Administration was performed once a day, five times a week, at a dose of 100 mg/kg for two weeks. Single administration groups and a combined administration group with the antibody-drug conjugate (1) and tucatinib, and a solvent administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (1) and tucatinib are shown in Figure 16 .
  • the TGI of single administration of tucatinib was 78%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 78%.
  • the tumor growth inhibition (TGI: 96%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • MDA-MB-453 cells a human breast cancer cell line, purchased from ATCC (American Type Culture Collection) were suspended in Matrigel basement membrane matrix, and 1 ⁇ 10 7 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0).
  • the antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 0.5 mg/kg on Day 0.
  • Tucatinib was administered once a day, five times a week, at a dose of 100 mg/kg for three weeks.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (1) and tucatinib, and a solvent administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (1) and tucatinib are shown in Figure 17 .
  • the TGI of single administration of tucatinib was 36%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 52%.
  • the tumor growth inhibition (TGI: 85%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • NCI-N87 cells a human gastric cancer cell line, purchased from ATCC (American Type Culture Collection) were suspended in physiological saline, and 1 ⁇ 10 7 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 6 days after the transplantation (Day 0).
  • the antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 2 mg/kg on Day 0.
  • Tucatinib was administered once a day, five times a week, at a dose of 100 mg/kg for two weeks.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (1) and tucatinib, and a solvent administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (1) and tucatinib are shown in Figure 18 .
  • the TGI of single administration of tucatinib was 65%.
  • the TGI of single administration of the antibody-drug conjugate (1) was 71%.
  • the tumor growth inhibition (TGI: 92%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • an anti-HER3 antibody an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4
  • KPL-4 cells a human breast cancer cell line, obtained from Dr. Junichi Kurebayashi, Kawasaki Medical School ( British Journal of Cancer. (1999)79 (5/6). 707-717 ) were suspended in physiological saline, and 1.5 ⁇ 10 7 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 17 days after the transplantation (Day 0).
  • the antibody-drug conjugate (2) was diluted with ABS buffer, and intravenously administered to the tail vein of each mouse once a week at a dose of 10 mg/kg (a liquid volume of 10 mL/kg) for three weeks (Days 0, 7, 14).
  • Neratinib was suspended in 0.5% methyl cellulose, and orally administered once a day, five times a week, at a dose of 20 mg/kg (a liquid volume of 10 mL/kg) for two weeks.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (2) and neratinib, and an ABS administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (2) and neratinib are shown in Figure 19 .
  • the TGI of single administration of neratinib on the day of effect determination was 84%.
  • the TGI of single administration of the antibody-drug conjugate (2) was 67%.
  • the tumor growth inhibition (TGI: 95%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • an anti-CDH6 antibody an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10
  • the antibody-drug conjugate (3) was intravenously administered to the tail vein of each mouse at a dose of 10 mg/kg on Day 0.
  • Cabozantinib was administered once a day, five times a week, 20 times in total, at a dose of 40 mg/kg.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (3) and cabozantinib, and a solvent administration group as a control group were established.
  • results of combined use of the antibody-drug conjugate (3) and cabozantinib are shown in Figure 20 .
  • the TGI of single administration of cabozantinib was 67%.
  • the TGI of single administration of the antibody-drug conjugate (3) was 71%.
  • the tumor growth inhibition (TGI: 90%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • an anti-TROP2 antibody an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6
  • erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with dimethyl sulfoxide (DMSO) to concentrations of 2 mM, 400 ⁇ M, 80 ⁇ M, 16 ⁇ M, 3.2 ⁇ M, and 640 nM, or DMSO was added at 25 nL/well.
  • DMSO dimethyl sulfoxide
  • Compound (1) prepared with an RPMI1640 Medium (Thermo Fisher Scientific) containing 10% fetal bovine serum (GE Healthcare) to concentrations of 60 nM, 24 nM, 9.6 nM, 3.8 nM, 1.5 nM, and 0.61 nM, or the antibody-drug conjugate (4) prepared to concentrations of 32 nM, 11 nM, 3.6 nM, 1.2 nM, 0.40 nM, and 0.13 nM, or the antibody-drug conjugate (1) prepared to concentrations of 8.0 nM, 2.7 nM, 0.89 nM, 0.30 nM, 0.10 nM, and 0.033 nM was added at 2.5 ⁇ L/well.
  • RPMI1640 Medium Thermo Fisher Scientific
  • 10% fetal bovine serum GE Healthcare
  • NCI-N87 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 4 ⁇ 10 4 cells/mL were seeded at 2.5 ⁇ L/well, and cultured at 37°C under 5%CO 2 for 6 days.
  • Cell growth inhibition rate (%) was calculated by using the following calculation expression.
  • Cell growth inhibition rate % 100 ⁇ T ⁇ B / C ⁇ B ⁇ 100
  • Synergy Score 0 indicates additive action
  • Synergy Score > 0 indicates synergistic action
  • Synergy Score ⁇ 0 indicates antagonistic action.
  • Synergy Scores for the combinations are shown in Table 1.
  • Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride) or brigatinib
  • the antibody-drug conjugate (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120
  • the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120.
  • the human breast cancer cell line KPL-4 obtained from Dr. Junichi Kurebayashi, Kawasaki Medical School ( British Journal of Cancer. (1999)79 (5/6). 707-717 ) was used for evaluation.
  • erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 ⁇ M, 80 ⁇ M, 16 ⁇ M, 3.2 ⁇ M, and 640 nM, or DMSO was added at 25 nL/well.
  • Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 36 nM, 20 nM, 11 nM, 6.2 nM, 3.4 nM, and 1.9 nM, or the antibody-drug conjugate (4) prepared to concentrations of 40 nM, 13 nM, 4.4 nM, 1.5 nM, 0.49 nM, and 0.16 nM, or the antibody-drug conjugate (1) prepared to concentrations of 3.7 nM, 1.3 nM, 0.48 nM, 0.17 nM, 0.061 nM, and 0.022 nM was added at 2.5 ⁇ L/well.
  • KPL-4 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 1 ⁇ 10 4 cells/mL were seeded at 2.5 ⁇ L/well, and cultured at 37°C under 5%CO 2 for 6 days. After culturing, a solution obtained by diluting CellTiter-Glo 2.0 Assay with an equivalent amount of Glo Lysis buffer, 1 ⁇ was added at 2 ⁇ L/well, and incubation was performed at room temperature for 1 hour, and luminescence intensity was then measured for each well.
  • Synergy Scores for the combinations are shown in Table 2.
  • Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), mubritinib, or BIBF1120
  • the antibody-drug conjugate (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120
  • the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), mubritinib, brigatinib, or BIBF1120.
  • the human lung cancer cell line EBC-1 obtained from Health Science Research Resources Bank (currently known as Japanese Collection of Research Bioresources (JCRB) Cell Bank) was used for evaluation.
  • erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 ⁇ M, 80 ⁇ M, 16 ⁇ M, 3.2 ⁇ M, and 640 nM or DMSO was added at 25 nL/well.
  • Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 16 nM, 8.0 nM, 4.0 nM, 2.0 nM, 1.0 nM, and 0.50 nM, or the antibody-drug conjugate (4) prepared to concentrations of 40 nM, 13 nM, 4.4 nM, 1.5 nM, 0.49 nM, and 0.16 nM was added at 2.5 ⁇ L/well. Subsequently, EBC-1 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 2 ⁇ 10 4 cells/mL were seeded at 2.5 ⁇ L/well, and cultured at 37°C under 5%CO 2 for 6 days.
  • Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120.
  • Example 17 Cell growth inhibition test (4)
  • the human breast cancer cell line HCC70 obtained from ATCC was used for evaluation.
  • erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 ⁇ M, 80 ⁇ M, 16 ⁇ M, 3.2 ⁇ M, and 640 nM or DMSO was added at 25 nL/well.
  • Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 800 nM, 200 nM, 50 nM, 13 nM, 3.1 nM, and 0.78 nM, or the antibody-drug conjugate (4) prepared to concentrations of 5.6 nM, 2.0 nM, 0.71 nM, 0.26 nM, 0.091 nM, and 0.033 nM was added at 2.5 ⁇ L/well. Subsequently, HCC70 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 4 ⁇ 10 4 cells/mL were seeded at 2.5 ⁇ L/well, and cultured at 37°C under 5%CO 2 for 6 days.
  • Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120.
  • the human pancreatic cancer cell line BxPC-3 obtained from ATCC was used for evaluation.
  • erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 ⁇ M, 80 ⁇ M, 16 ⁇ M, 3.2 ⁇ M, and 640 nM or DMSO was added at 25 nL/well.
  • Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 80 nM, 32 nM, 13 nM, 5.1 nM, 2.0 nM, and 0.82 nM, or the antibody-drug conjugate (4) prepared to concentrations of 16 nM, 5.3 nM, 1.8 nM, 0.59 nM, 0.20 nM, and 0.066 nM was added at 2.5 ⁇ L/well. Subsequently, BxPC-3 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 4 ⁇ 10 4 cells/mL were seeded at 2.5 ⁇ L/well, and cultured at 37°C under 5%CO 2 for 6 days.
  • Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120.
  • MDA-MB-453 cells a human breast cancer cell line, obtained from ATCC were suspended in Matrigel basement membrane matrix, and 1 ⁇ 10 7 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 11 days after the transplantation (Day 0).
  • the antibody-drug conjugate (2) was diluted with ABS buffer, and intravenously administered to the tail vein of each mouse once a week at a dose of 1 mg/kg (a liquid volume of 10 mL/kg) for three weeks (Days 0, 7, 14).
  • Palbociclib was suspended in 0.5% methyl cellulose, and orally administered once a day, five times a week, at a dose of 50 mg/kg (a liquid volume of 10 mL/kg) for three weeks from Day 0.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (2) and palbociclib, and an ABS administration group as a control group were established.
  • TGI of single administration of palbociclib was 72%.
  • the TGI of single administration of the antibody-drug conjugate (2) was 54%.
  • the tumor growth inhibition (TGI: 93%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • MDA-MB-453 cells a human breast cancer cell line, obtained from ATCC were suspended in Matrigel basement membrane matrix, and 1 ⁇ 10 7 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0).
  • the antibody-drug conjugate (2) was diluted with ABS buffer, and intravenously administered to the tail vein of each mouse once a week at a dose of 1 mg/kg (a liquid volume of 10 mL/kg) for three weeks (Days 0, 7, 14).
  • Alpelisib was suspended in 0.5% methyl cellulose, and orally administered once a day, five times a week, at a dose of 25 mg/kg (a liquid volume of 10 mL/kg) for three weeks from Day 0.
  • Single administration groups and a combined administration group with the antibody-drug conjugate (2) and alpelisib, and an ABS administration group as a control group were established.
  • TGI of single administration of alpelisib was 56%.
  • TGI of single administration of the antibody-drug conjugate (2) was 68%.
  • the tumor growth inhibition (TGI: 86%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A pharmaceutical composition wherein an antibody-drug conjugate in which a drug-linker represented by the following formula (wherein A represents a connecting position to an antibody) is conjugated to the antibody via a thioether bond and a kinase inhibitor (at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor) are administered in combination, and/or a method of treatment wherein the antibody-drug conjugate and a kinase inhibitor are administered in combination to a subject.
Figure imga0001

Description

    Technical Field
  • The present invention relates to a pharmaceutical composition wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination, and/or a method of treatment wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination to a subject.
  • Background Art
  • Abnormal activation of intracellular signaling is associated with proliferation of cancer cells and tumor angiogenesis. Kinase inhibitors are agents that inhibit kinases involved in abnormally activated intracellular signaling to exert an antitumor effect. Examples of such kinase inhibitors include a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, an PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, and an NTRK inhibitor (Non-Patent References 1 to 11).
  • An antibody-drug conjugate (ADC) having a drug with cytotoxicity conjugated to an antibody, whose antigen is expressed on the surface of cancer cells and which also binds to an antigen capable of cellular internalization, and therefore can deliver the drug selectively to cancer cells, is thus expected to cause accumulation of the drug within cancer cells and to kill the cancer cells (Non-Patent References 12 to 16).
  • As one such antibody-drug conjugate, an antibody-drug conjugate comprising an antibody and a derivative of exatecan, which is a topoisomerase I inhibitor, as its components is known (Patent References 1 to 8, Non-Patent References 17 to 21).
  • Patent References 1 to 8 disclose that the antibody-drug conjugate can be administered together with various cancer treating agents.
  • However, none of the references describes any test result showing an excellent combined effect when the foregoing antibody-drug conjugate and the kinase inhibitor are used in combination, or any scientific basis for suggesting such a test result.
  • Citation List Patent Literature
  • Non-Patent Literature
    • Non-Patent Reference 1: Otto T., et al., Nat. Rev. Cancer (2017) 17(2) : 93-115.
    • Non-Patent Reference 2: Zhang YJ., et al., Drug Discov. Today (2011) 16(7-8) : 325-331.
    • Non-Patent Reference 3: Zaytseva YY., et al., Cancer Lett. (2012) 319(1) : 1-7.
    • Non-Patent Reference 4: Janku F., et al., Nat. Rev. Clin. Oncol. (2018) 15(5): 273-291.
    • Non-Patent Reference 5: Bergholz JS., et al., J. Clin. Oncol. (2018) 36(13): 1339-1342
    • Non-Patent Reference 6: Zhao Y., et al., Nat. Rev. Clin. Oncol. (2014) 11(7): 385-400.
    • Non-Patent Reference 7: Caunt CJ., et al., Nat. Rev. Cancer (2015) 15(10): 577-592.
    • Non-Patent Reference 8: Ryan MB., et al., Nat. Rev. Clin. Oncol. (2018) 15(11): 709-720.
    • Non-Patent Reference 9: Ferquson FM., et al., Nat. Rev. Drug Discov. (2018) 17(5): 353-377.
    • Non-Patent Reference 10: Konig H., et al., Current Cancer Drug Targets (2015) 15, 803-821.
    • Non-Patent Reference 11: Kheder ES., et al., Clinical Cancer Research (2018) 24(23), 5807-5814.
    • Non-Patent Reference 12: Ducry, L., et al., Bioconjugate Chem. (2010) 21, 5-13.
    • Non-Patent Reference 13: Alley, S. C., et al., Current Opinion in Chemical Biology (2010) 14, 529-537.
    • Non-Patent Reference 14: Damle N. K. Expert Opin. Biol. Ther. (2004) 4, 1445-1452.
    • Non-Patent Reference 15: Senter P. D., et al., Nature Biotechnology (2012) 30, 631-637.
    • Non-Patent Reference 16: Burris HA., et al., J. Clin. Oncol. (2011) 29(4): 398-405.
    • Non-Patent Reference 17: Ogitani Y. et al., Clinical Cancer Research (2016) 22(20), 5097-5108.
    • Non-Patent Reference 18: Ogitani Y. et al., Cancer Science (2016) 107, 1039-1046.
    • Non-Patent Reference 19: Doi T, et al., Lancet Oncol. (2017) 18, 1512-22.
    • Non-Patent Reference 20: Takegawa N, et al., Int. J. Cancer (2017) 141, 1682-1689.
    • Non-Patent Reference 21: Yonesaka K, et al., Int. Oncogene (2018) 141, 1682-1689 (2017).
    Summary of Invention Technical Problem
  • An antibody-drug conjugate used in the present invention (an antibody-drug conjugate including a derivative of exatecan as a component) has been confirmed to exert a superior antitumor effect even as a single agent. However, there has been a need to obtain a method of treatment which can suppress growth of cancer cells in multiple manners and exert a further superior antitumor effect by using the antibody-drug conjugate in combination with another anticancer agent having a different mechanism of action.
  • An object of the present invention is to provide a pharmaceutical composition wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination, and/or a method of treatment wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination to a subject.
  • Solution to Problem
  • As a result of diligent studies in order to solve the above problems, the present inventors have found that combined administration of a specific antibody-drug conjugate and a kinase inhibitor exhibits a superior combined effect, and completed the present invention.
  • Thus, the present invention provides the following [1] to [1072].
    1. [1] A pharmaceutical composition wherein an antibody-drug conjugate and a kinase inhibitor are administered in combination, and the antibody-drug conjugate is an antibody-drug conjugate in which a drug-linker represented by the following formula:
      Figure imgb0001
      Figure imgb0002
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    2. [2] The pharmaceutical composition according to [1], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    3. [3] The pharmaceutical composition according to [2], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    4. [4] The pharmaceutical composition according to [3], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    5. [5] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an mTOR inhibitor.
    6. [6] The pharmaceutical composition according to [5], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    7. [7] The pharmaceutical composition according to [2], wherein the kinase inhibitor is a PI3K inhibitor.
    8. [8] The pharmaceutical composition according to [7], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    9. [9] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an RAF inhibitor.
    10. [10] The pharmaceutical composition according to [9], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    11. [11] The pharmaceutical composition according to [2], wherein the kinase inhibitor is a VEGFR inhibitor.
    12. [12] The pharmaceutical composition according to [11], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    13. [13] The pharmaceutical composition according to [2], wherein the kinase inhibitor is a KIT inhibitor.
    14. [14] The pharmaceutical composition according to [13], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    15. [15] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an RET inhibitor.
    16. [16] The pharmaceutical composition according to [15], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    17. [17] The pharmaceutical composition according to [2], wherein the kinase inhibitor is a PDGFR inhibitor.
    18. [18] The pharmaceutical composition according to [17], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    19. [19] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an FGFR inhibitor.
    20. [20] The pharmaceutical composition according to [19], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    21. [21] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an FLT3 inhibitor.
    22. [22] The pharmaceutical composition according to [21], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    23. [23] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an ALK inhibitor.
    24. [24] The pharmaceutical composition according to [23], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    25. [25] The pharmaceutical composition according to [2], wherein the kinase inhibitor is a CSF-1R inhibitor.
    26. [26] The pharmaceutical composition according to [25], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    27. [27] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an EGFR inhibitor.
    28. [28] The pharmaceutical composition according to [27], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    29. [29] The pharmaceutical composition according to [2], wherein the kinase inhibitor is an HER2 inhibitor.
    30. [30] The pharmaceutical composition according to [29], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    31. [31] The pharmaceutical composition according to any one of [1] to [30], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    32. [32] The pharmaceutical composition according to [31], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    33. [33] The pharmaceutical composition according to [32], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    34. [34] The pharmaceutical composition according to [32], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    35. [35] The pharmaceutical composition according to [32], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    36. [36] The pharmaceutical composition according to [32], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    37. [37] The pharmaceutical composition according to any one of [32] to [36], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    38. [38] The pharmaceutical composition according to [31], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    39. [39] The pharmaceutical composition according to [38], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    40. [40] The pharmaceutical composition according to [39], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    41. [41] The pharmaceutical composition according to any one of [38] to [40], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    42. [42] The pharmaceutical composition according to [31], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    43. [43] The pharmaceutical composition according to [42], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    44. [44] The pharmaceutical composition according to [43], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    45. [45] The pharmaceutical composition according to any one of [42] to [44], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    46. [46] The pharmaceutical composition according to [31], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    47. [47] The pharmaceutical composition according to [46], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    48. [48] The pharmaceutical composition according to [47], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    49. [49] The pharmaceutical composition according to any one of [46] to [48], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    50. [50] The pharmaceutical composition according to [31], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    51. [51] The pharmaceutical composition according to [50], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    52. [52] The pharmaceutical composition according to [51], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    53. [53] The pharmaceutical composition according to any one of [50] to [52], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    54. [54] The pharmaceutical composition according to any one of [1] to [53], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    55. [55] The pharmaceutical composition according to any one of [1] to [54], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    56. [56] The pharmaceutical composition according to [55], wherein the pharmaceutical composition is for use in treating breast cancer.
    57. [57] The pharmaceutical composition according to [55], wherein the pharmaceutical composition is for use in treating colorectal cancer.
    58. [58] The pharmaceutical composition according to [55], wherein the pharmaceutical composition is for use in treating gastric cancer.
    59. [59] The pharmaceutical composition according to [55], wherein the pharmaceutical composition is for use in treating lung cancer.
    60. [60] The pharmaceutical composition according to [55], wherein the pharmaceutical composition is for use in treating pancreatic cancer.
    61. [61] The pharmaceutical composition according to [55], wherein the pharmaceutical composition is for use in treating kidney cancer.
    62. [62] The pharmaceutical composition according to [55], wherein the pharmaceutical composition is for use in treating ovarian cancer.
    63. [63] A pharmaceutical composition wherein an antibody-drug conjugate and a kinase inhibitor are administered in combination, and the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0003
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    64. [64] The pharmaceutical composition according to [63], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    65. [65] The pharmaceutical composition according to [64], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    66. [66]The pharmaceutical composition according to [65], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    67. [67] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an mTOR inhibitor.
    68. [68] The pharmaceutical composition according to [67], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    69. [69] The pharmaceutical composition according to [64], wherein the kinase inhibitor is a PI3K inhibitor.
    70. [70] The pharmaceutical composition according to [69], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    71. [71] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an RAF inhibitor.
    72. [72] The pharmaceutical composition according to [71], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    73. [73] The pharmaceutical composition according to [64], wherein the kinase inhibitor is a VEGFR inhibitor.
    74. [74] The pharmaceutical composition according to [73], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    75. [75] The pharmaceutical composition according to [64], wherein the kinase inhibitor is a KIT inhibitor.
    76. [76] The pharmaceutical composition according to [75], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    77. [77] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an RET inhibitor.
    78. [78] The pharmaceutical composition according to [77], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    79. [79] The pharmaceutical composition according to [64], wherein the kinase inhibitor is a PDGFR inhibitor.
    80. [80] The pharmaceutical composition according to [79], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    81. [81] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an FGFR inhibitor.
    82. [82] The pharmaceutical composition according to [81], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    83. [83] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an FLT3 inhibitor.
    84. [84] The pharmaceutical composition according to [83], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    85. [85] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an ALK inhibitor.
    86. [86] The pharmaceutical composition according to [85], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    87. [87] The pharmaceutical composition according to [64], wherein the kinase inhibitor is a CSF-1R inhibitor.
    88. [88] The pharmaceutical composition according to [87], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    89. [89] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an EGFR inhibitor.
    90. [90] The pharmaceutical composition according to [89], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    91. [91] The pharmaceutical composition according to [64], wherein the kinase inhibitor is an HER2 inhibitor.
    92. [92] The pharmaceutical composition according to [91], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    93. [93] The pharmaceutical composition according to any one of [63] to [92], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    94. [94] The pharmaceutical composition according to [93], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    95. [95] The pharmaceutical composition according to [94], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    96. [96] The pharmaceutical composition according to [94], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    97. [97] The pharmaceutical composition according to [94], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    98. [98] The pharmaceutical composition according to [94], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    99. [99] The pharmaceutical composition according to any one of [94] to [98], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    100. [100] The pharmaceutical composition according to [93], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    101. [101] The pharmaceutical composition according to [100], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    102. [102] The pharmaceutical composition according to [101], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    103. [103] The pharmaceutical composition according to any one of [100] to [102], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    104. [104] The pharmaceutical composition according to [93], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    105. [105] The pharmaceutical composition according to [104], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    106. [106] The pharmaceutical composition according to [105], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    107. [107] The pharmaceutical composition according to any one of [104] to [106], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    108. [108] The pharmaceutical composition according to [93], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    109. [109] The pharmaceutical composition according to [108], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    110. [110] The pharmaceutical composition according to [109], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    111. [111] The pharmaceutical composition according to any one of [108] to [110], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    112. [112] The pharmaceutical composition according to [93], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    113. [113] The pharmaceutical composition according to [112], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    114. [114] The pharmaceutical composition according to [113], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    115. [115] The pharmaceutical composition according to any one of [112] to [114], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    116. [116] The pharmaceutical composition according to any one of [63] to [115], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    117. [117] The pharmaceutical composition according to any one of [63] to [116], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    118. [118] The pharmaceutical composition according to [117], wherein the pharmaceutical composition is for use in treating breast cancer.
    119. [119] The pharmaceutical composition according to [117], wherein the pharmaceutical composition is for use in treating colorectal cancer.
    120. [120] The pharmaceutical composition according to [117], wherein the pharmaceutical composition is for use in treating gastric cancer.
    121. [121] The pharmaceutical composition according to [117], wherein the pharmaceutical composition is for use in treating lung cancer.
    122. [122] The pharmaceutical composition according to [117], wherein the pharmaceutical composition is for use in treating pancreatic cancer.
    123. [123] The pharmaceutical composition according to [117], wherein the pharmaceutical composition is for use in treating kidney cancer.
    124. [124] The pharmaceutical composition according to [117], wherein the pharmaceutical composition is for use in treating ovarian cancer.
    125. [125] A method of treatment, comprising administering an antibody-drug conjugate and a kinase inhibitor in combination to a subject in need of treatment, wherein the antibody-drug conjugate is an antibody-drug conjugate in which a drug-linker represented by the following formula:
      Figure imgb0004
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    126. [126] The method of treatment according to [125], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    127. [127] The method of treatment according to [126], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    128. [128] The method of treatment according to [127], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    129. [129] The method of treatment according to [126], wherein the kinase inhibitor is an mTOR inhibitor.
    130. [130] The method of treatment according to [129], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    131. [131] The method of treatment according to [126], wherein the kinase inhibitor is a PI3K inhibitor.
    132. [132] The method of treatment according to [131], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    133. [133] The method of treatment according to [126], wherein the kinase inhibitor is an RAF inhibitor.
    134. [134] The method of treatment according to [133], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    135. [135] The method of treatment according to [126], wherein the kinase inhibitor is a VEGFR inhibitor.
    136. [136] The method of treatment according to [135], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    137. [137] The method of treatment according to [126], wherein the kinase inhibitor is a KIT inhibitor.
    138. [138] The method of treatment according to [137], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    139. [139] The method of treatment according to [126], wherein the kinase inhibitor is an RET inhibitor.
    140. [140] The method of treatment according to [139], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    141. [141] The method of treatment according to [126], wherein the kinase inhibitor is a PDGFR inhibitor.
    142. [142] The method of treatment according to [141], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    143. [143] The method of treatment according to [126], wherein the kinase inhibitor is a FGFR inhibitor.
    144. [144] The method of treatment according to [143], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    145. [145] The method of treatment according to [126], wherein the kinase inhibitor is a FLT3 inhibitor.
    146. [146] The method of treatment according to [145], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    147. [147] The method of treatment according to [126], wherein the kinase inhibitor is an ALK inhibitor.
    148. [148] The method of treatment according to [147], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    149. [149] The method of treatment according to [126], wherein the kinase inhibitor is a CSF-1R inhibitor.
    150. [150] The method of treatment according to [149], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    151. [151] The method of treatment according to [126], wherein the kinase inhibitor is an EGFR inhibitor.
    152. [152] The method of treatment according to [151], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    153. [153] The method of treatment according to [126], wherein the kinase inhibitor is an HER2 inhibitor.
    154. [154] The method of treatment according to [153], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    155. [155] The method of treatment according to any one of [125] to [154], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    156. [156] The method of treatment according to [155], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    157. [157] The method of treatment according to [156], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    158. [158] The method of treatment according to [156], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    159. [159] The method of treatment according to [156], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    160. [160] The method of treatment according to [156], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    161. [161] The method of treatment according to any one of [156] to [160], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    162. [162] The method of treatment according to [155], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    163. [163] The method of treatment according to [162], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    164. [164] The method of treatment according to [163], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    165. [165] The method of treatment according to any one of [162] to [164], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    166. [166] The method of treatment according to [155], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    167. [167] The method of treatment according to [166], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    168. [168] The method of treatment according to [167], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    169. [169] The method of treatment according to any one of [166] to [168], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    170. [170] The method of treatment according to [155], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    171. [171] The method of treatment according to [170], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    172. [172] The method of treatment according to [171], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    173. [173] The method of treatment according to any one of [170] to [172], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    174. [174] The method of treatment according to [155], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    175. [175] The method of treatment according to [174], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    176. [176] The method of treatment according to [175], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    177. [177] The method of treatment according to any one of [174] to [176], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    178. [178] The method of treatment according to any one of [125] to [177], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    179. [179] The method of treatment according to any one of [125] to [178], wherein the method of treatment is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    180. [180] The method of treatment according to [179], wherein the method of treatment is for treating breast cancer.
    181. [181] The method of treatment according to [179], wherein the method of treatment is for treating colorectal cancer.
    182. [182] The method of treatment according to [179], wherein the method of treatment is for treating gastric cancer.
    183. [183] The method of treatment according to [179], wherein the method of treatment is for treating lung cancer.
    184. [184] The method of treatment according to [179], wherein the method of treatment is for treating pancreatic cancer.
    185. [185] The method of treatment according to [179], wherein the method of treatment is for treating kidney cancer.
    186. [186] The method of treatment according to [179], wherein the method of treatment is for treating ovarian cancer.
    187. [187] A method of treatment, comprising administering an antibody-drug conjugate and a kinase inhibitor in combination to a subject in need of treatment, wherein the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0005
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    188. [188] The method of treatment according to [187], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    189. [189] The method of treatment according to [188], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    190. [190] The method of treatment according to [189], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    191. [191] The method of treatment according to [188], wherein the kinase inhibitor is an mTOR inhibitor.
    192. [192] The method of treatment according to [191], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    193. [193] The method of treatment according to [188], wherein the kinase inhibitor is a PI3K inhibitor.
    194. [194] The method of treatment according to [193], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    195. [195] The method of treatment according to [188], wherein the kinase inhibitor is an RAF inhibitor.
    196. [196] The method of treatment according to [195], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    197. [197] The method of treatment according to [188], wherein the kinase inhibitor is a VEGFR inhibitor.
    198. [198] The method of treatment according to [197], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    199. [199] The method of treatment according to [188], wherein the kinase inhibitor is a KIT inhibitor.
    200. [200] The method of treatment according to [199], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    201. [201] The method of treatment according to [188], wherein the kinase inhibitor is an RET inhibitor.
    202. [202] The method of treatment according to [201], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    203. [203] The method of treatment according to [188], wherein the kinase inhibitor is a PDGFR inhibitor.
    204. [204] The method of treatment according to [203], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    205. [205] The method of treatment according to [188], wherein the kinase inhibitor is a FGFR inhibitor.
    206. [206] The method of treatment according to [205], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    207. [207] The method of treatment according to [188], wherein the kinase inhibitor is an FLT3 inhibitor.
    208. [208] The method of treatment according to [207], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    209. [209] The method of treatment according to [188], wherein the kinase inhibitor is an ALK inhibitor.
    210. [210] The method of treatment according to [209], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    211. [211] The method of treatment according to [188], wherein the kinase inhibitor is a CSF-1R inhibitor.
    212. [212] The method of treatment according to [211], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    213. [213] The method of treatment according to [188], wherein the kinase inhibitor is an EGFR inhibitor.
    214. [214] The method of treatment according to [213], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    215. [215] The method of treatment according to [188], wherein the kinase inhibitor is an HER2 inhibitor.
    216. [216] The method of treatment according to [215], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    217. [217] The method of treatment according to any one of [187] to [216], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    218. [218] The method of treatment according to [217], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    219. [219] The method of treatment according to [218], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    220. [220] The method of treatment according to [218], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    221. [221] The method of treatment according to [218], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    222. [222] The method of treatment according to [218], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    223. [223] The method of treatment according to any one of [218] to [222], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    224. [224] The method of treatment according to [217], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    225. [225] The method of treatment according to [224], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    226. [226] The method of treatment according to [225], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    227. [227] The method of treatment according to any one of [224] to [226], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    228. [228] The method of treatment according to [217], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    229. [229] The method of treatment according to [228], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    230. [230] The method of treatment according to [229], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    231. [231] The method of treatment according to any one of [228] to [230], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    232. [232] The method of treatment according to [217], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    233. [233] The method of treatment according to [232], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    234. [234] The method of treatment according to [233], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    235. [235] The method of treatment according to any one of [232] to [234], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    236. [236] The method of treatment according to [217], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    237. [237] The method of treatment according to [236], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    238. [238] The method of treatment according to [237], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    239. [239] The method of treatment according to any one of [236] to [238], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    240. [240] The method of treatment according to any one of [187] to [231], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    241. [241] The method of treatment according to any one of [187] to [240], wherein the method of treatment is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    242. [242] The method of treatment according to [241], wherein the method of treatment is for treating breast cancer.
    243. [243] The method of treatment according to [241], wherein the method of treatment is for treating colorectal cancer.
    244. [244] The method of treatment according to [241], wherein the method of treatment is for treating gastric cancer.
    245. [245] The method of treatment according to [241], wherein the method of treatment is for treating lung cancer.
    246. [246] The method of treatment according to [241], wherein the method of treatment is for treating pancreatic cancer.
    247. [247] The method of treatment according to [241], wherein the method of treatment is for treating kidney cancer.
    248. [248] The method of treatment according to [241], wherein the method of treatment is for treating ovarian cancer.
    249. [249] An antibody-drug conjugate for use in treating a disease through being administered in combination with a kinase inhibitor, wherein a drug-linker represented by the following formula:
      Figure imgb0006
      Figure imgb0007
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond in the antibody-drug conjugate, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    250. [250] The antibody-drug conjugate according to [249], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    251. [251] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    252. [252] The antibody-drug conjugate according to [251], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    253. [253] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an mTOR inhibitor.
    254. [254] The antibody-drug conjugate according to [253], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    255. [255] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is a PI3K inhibitor.
    256. [256] The antibody-drug conjugate according to [255], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    257. [257] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an RAF inhibitor.
    258. [258] The antibody-drug conjugate according to [257], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    259. [259] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is a VEGFR inhibitor.
    260. [260] The antibody-drug conjugate according to [259], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    261. [261] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is a KIT inhibitor.
    262. [262] The antibody-drug conjugate according to [261], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    263. [263] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an RET inhibitor.
    264. [264] The antibody-drug conjugate according to [263], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    265. [265] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is a PDGFR inhibitor.
    266. [266] The antibody-drug conjugate according to [265], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    267. [267] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an FGFR inhibitor.
    268. [268] The antibody-drug conjugate according to [267], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    269. [269] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an FLT3 inhibitor.
    270. [270] The antibody-drug conjugate according to [269], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    271. [271] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an ALK inhibitor.
    272. [272] The antibody-drug conjugate according to [271], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    273. [273] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is a CSF-1R inhibitor.
    274. [274] The antibody-drug conjugate according to [273], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    275. [275] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an EGFR inhibitor.
    276. [276] The antibody-drug conjugate according to [275], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    277. [277] The antibody-drug conjugate according to [250], wherein the kinase inhibitor is an HER2 inhibitor.
    278. [278] The antibody-drug conjugate according to [277], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    279. [279] The antibody-drug conjugate according to any one of [249] to [278], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    280. [280] The antibody-drug conjugate according to [279], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    281. [281] The antibody-drug conjugate according to [280], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    282. [282] The antibody-drug conjugate according to [280], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    283. [283] The antibody-drug conjugate according to [280], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    284. [284] The antibody-drug conjugate according to [280], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    285. [285] The antibody-drug conjugate according to any one of [280] to [284], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    286. [286] The antibody-drug conjugate according to [279], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    287. [287] The antibody-drug conjugate according to [286], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    288. [288] The antibody-drug conjugate according to [287], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    289. [289] The antibody-drug conjugate according to any one of [286] to [288], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    290. [290] The antibody-drug conjugate according to [279], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    291. [291] The antibody-drug conjugate according to [290], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    292. [292] The antibody-drug conjugate according to [291], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    293. [293] The antibody-drug conjugate according to any one of [290] to [292], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    294. [294] The antibody-drug conjugate according to [279], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    295. [295] The antibody-drug conjugate according to [294], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    296. [296] The antibody-drug conjugate according to [295], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    297. [297] The antibody-drug conjugate according to any one of [294] to [296], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    298. [298] The antibody-drug conjugate according to [279], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    299. [299] The antibody-drug conjugate according to [298], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    300. [300] The antibody-drug conjugate according to [299], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    301. [301] The antibody-drug conjugate according to any one of [298] to [300], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    302. [302] The antibody-drug conjugate according to any one of [249] to [301], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    303. [303] The antibody-drug conjugate according to any one of [249] to [302], wherein the antibody-drug conjugate is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    304. [304] The antibody-drug conjugate according to [303], wherein the antibody-drug conjugate is for use in treating breast cancer.
    305. [305] The antibody-drug conjugate according to [303], wherein the antibody-drug conjugate is for use in treating colorectal cancer.
    306. [306] The antibody-drug conjugate according to [303], wherein the antibody-drug conjugate is for use in treating gastric cancer.
    307. [307] The antibody-drug conjugate according to [303], wherein the antibody-drug conjugate is for use in treating lung cancer.
    308. [308] The antibody-drug conjugate according to [303], wherein the antibody-drug conjugate is for use in treating pancreatic cancer.
    309. [309] The antibody-drug conjugate according to [303], wherein the antibody-drug conjugate is for use in treating kidney cancer.
    310. [310] The antibody-drug conjugate according to [303], wherein the antibody-drug conjugate is for use in treating ovarian cancer.
    311. [311] An antibody-drug conjugate for use in treating a disease through being administered in combination with a kinase inhibitor, wherein the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0008
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    312. [312] The antibody-drug conjugate according to [311], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    313. [313] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    314. [314] The antibody-drug conjugate according to [313], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    315. [315] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an mTOR inhibitor.
    316. [316] The antibody-drug conjugate according to [315], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    317. [317] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is a PI3K inhibitor.
    318. [318] The antibody-drug conjugate according to [317], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    319. [319] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an RAF inhibitor.
    320. [320] The antibody-drug conjugate according to [319], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    321. [321] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is a VEGFR inhibitor.
    322. [322] The antibody-drug conjugate according to [321], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    323. [323] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is a KIT inhibitor.
    324. [324] The antibody-drug conjugate according to [323], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    325. [325] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an RET inhibitor.
    326. [326] The antibody-drug conjugate according to [325], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    327. [327] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is a PDGFR inhibitor.
    328. [328] The antibody-drug conjugate according to [327], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    329. [329] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an FGFR inhibitor.
    330. [330] The antibody-drug conjugate according to [329], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    331. [331] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an FLT3 inhibitor.
    332. [332] The antibody-drug conjugate according to [331], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    333. [333] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an ALK inhibitor.
    334. [334] The antibody-drug conjugate according to [333], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    335. [335] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is a CSF-1R inhibitor.
    336. [336] The antibody-drug conjugate according to [335], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    337. [337] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an EGFR inhibitor.
    338. [338] The antibody-drug conjugate according to [337], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    339. [339] The antibody-drug conjugate according to [312], wherein the kinase inhibitor is an HER2 inhibitor.
    340. [340] The antibody-drug conjugate according to [339], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    341. [341] The antibody-drug conjugate according to any one of [311] to [340], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    342. [342] The antibody-drug conjugate according to [341], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    343. [343] The antibody-drug conjugate according to [342], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    344. [344] The antibody-drug conjugate according to [342], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    345. [345] The antibody-drug conjugate according to [342], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    346. [346] The antibody-drug conjugate according to [342], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    347. [347] The antibody-drug conjugate according to any one of [342] to [346], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    348. [348] The antibody-drug conjugate according to [341], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    349. [349] The antibody-drug conjugate according to [348], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    350. [350] The antibody-drug conjugate according to [349], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    351. [351] The antibody-drug conjugate according to any one of [348] to [350], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    352. [352] The antibody-drug conjugate according to [341], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    353. [353] The antibody-drug conjugate according to [352], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    354. [354] The antibody-drug conjugate according to [353], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    355. [355] The antibody-drug conjugate according to any one of [352] to [354], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    356. [356] The antibody-drug conjugate according to [341], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    357. [357] The antibody-drug conjugate according to [356], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    358. [358] The antibody-drug conjugate according to [357], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    359. [359] The antibody-drug conjugate according to any one of [356] to [358], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    360. [360] The antibody-drug conjugate according to [341], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    361. [361] The antibody-drug conjugate according to [360], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    362. [362] The antibody-drug conjugate according to [361], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    363. [363] The antibody-drug conjugate according to any one of [360] to [362], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    364. [364] The antibody-drug conjugate according to any one of [311] to [363], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    365. [365] The antibody-drug conjugate according to any one of [311] to [364], wherein the antibody-drug conjugate is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    366. [366] The antibody-drug conjugate according to [365], wherein the antibody-drug conjugate is for use in treating breast cancer.
    367. [367] The antibody-drug conjugate according to [365], wherein the antibody-drug conjugate is for use in treating colorectal cancer.
    368. [368] The antibody-drug conjugate according to [365], wherein the antibody-drug conjugate is for use in treating gastric cancer.
    369. [369] The antibody-drug conjugate according to [365], wherein the antibody-drug conjugate is for use in treating lung cancer.
    370. [370] The antibody-drug conjugate according to [365], wherein the antibody-drug conjugate is for use in treating pancreatic cancer.
    371. [371] The antibody-drug conjugate according to [365], wherein the antibody-drug conjugate is for use in treating kidney cancer.
    372. [372] The antibody-drug conjugate according to [365], wherein the antibody-drug conjugate is for use in treating ovarian cancer.
    373. [373] Use of an antibody-drug conjugate for the manufacture of a medicament for treating a disease through being administered in combination with a kinase inhibitor, wherein a drug-linker represented by the following formula:
      Figure imgb0009
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond in the antibody-drug conjugate, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    374. [374] The use according to [373], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    375. [375] The use according to [374], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    376. [376] The use according to [375], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    377. [377] The use according to [374], wherein the kinase inhibitor is an mTOR inhibitor.
    378. [378] The use according to [377], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    379. [379] The use according to [374], wherein the kinase inhibitor is a PI3K inhibitor.
    380. [380] The use according to [379], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    381. [381] The use according to [374], wherein the kinase inhibitor is an RAF inhibitor.
    382. [382] The use according to [381], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    383. [383] The use according to [374], wherein the kinase inhibitor is a VEGFR inhibitor.
    384. [384] The use according to [383], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    385. [385] The use according to [374], wherein the kinase inhibitor is a KIT inhibitor.
    386. [386] The use according to [385], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    387. [387] The use according to [374], wherein the kinase inhibitor is an RET inhibitor.
    388. [388] The use according to [387], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    389. [389] The use according to [374], wherein the kinase inhibitor is a PDGFR inhibitor.
    390. [390] The use according to [389], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    391. [391] The use according to [374], wherein the kinase inhibitor is an FGFR inhibitor.
    392. [392] The use according to [391], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    393. [393] The use according to [374], wherein the kinase inhibitor is an FLT3 inhibitor.
    394. [394] The use according to [393], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    395. [395] The use according to [374], wherein the kinase inhibitor is an ALK inhibitor.
    396. [396] The use according to [395], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    397. [397] The use according to [374], wherein the kinase inhibitor is a CSF-1R inhibitor.
    398. [398] The use according to [397], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    399. [399] The use according to [374], wherein the kinase inhibitor is an EGFR inhibitor.
    400. [400] The use according to [399], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    401. [401] The use according to [374], wherein the kinase inhibitor is an HER2 inhibitor.
    402. [402] The use according to [401], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    403. [403] The use according to any one of [373] to [402], wherein the antibody in the use is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    404. [404] The use according to [403], wherein the antibody in the use is an anti-HER2 antibody.
    405. [405] The use according to [404], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    406. [406] The use according to [404], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    407. [407] The use according to [404], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    408. [408] The use according to [404], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    409. [409] The use according to any one of [404] to [408], wherein the average number of units of the drug-linker conjugated per antibody molecule in the use is in the range of from 7 to 8.
    410. [410] The use according to [403], wherein the antibody in the use is an anti-HER3 antibody.
    411. [411] The use according to [410], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    412. [412] The use according to [411], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    413. [413] The use according to any one of [410] to [412], wherein the average number of units of the drug-linker conjugated per antibody molecule in the use is in the range of from 7 to 8.
    414. [414] The use according to [403], wherein the antibody in the use is an anti-TROP2 antibody.
    415. [415] The use according to [414], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    416. [416] The use according to [415], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    417. [417] The use according to any one of [414] to [416], wherein the average number of units of the drug-linker conjugated per antibody molecule in the use is in the range of from 3.5 to 4.5.
    418. [418] The use according to [403], wherein the antibody in the use is an anti-B7-H3 antibody.
    419. [419] The use according to [418], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    420. [420] The use according to [419], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    421. [421] The use according to any one of [418] to [420], wherein the average number of units of the drug-linker conjugated per antibody molecule in the use is in the range of from 3.5 to 4.5.
    422. [422] The use according to [403], wherein the antibody in the use is an anti-CDH6 antibody.
    423. [423] The use according to [422], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    424. [424] The use according to [423], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    425. [425] The use according to any one of [422] to [424], wherein the average number of units of the drug-linker conjugated per antibody molecule in the use is in the range of from 7 to 8.
    426. [426] The use according to any one of [373] to [425], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    427. [427] The use according to any one of [373] to [426], wherein the use is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroespophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    428. [428] The use according to [427], wherein the use is for treating breast cancer.
    429. [429] The use according to [427], wherein the use is for treating colorectal cancer.
    430. [430] The use according to [427], wherein the use is for treating gastric cancer.
    431. [431] The use according to [427], wherein the use is for treating lung cancer.
    432. [432] The use according to [427], wherein the use is for treating pancreatic cancer.
    433. [433] The use according to [427], wherein the use is for treating kidney cancer.
    434. [434] The use according to [427], wherein the use is for treating ovarian cancer.
    435. [435] Use of an antibody-drug conjugate for the manufacture of a medicament for treating a disease through being administered in combination with a kinase inhibitor, wherein the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0010
      Figure imgb0011
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    436. [436] The use according to [435], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    437. [437] The use according to [436], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    438. [438] The use according to [437], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    439. [439] The use according to [436], wherein the kinase inhibitor is an mTOR inhibitor.
    440. [440] The use according to [439], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    441. [441] The use according to [436], wherein the kinase inhibitor is a PI3K inhibitor.
    442. [442] The use according to [441], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    443. [443] The use according to [436], wherein the kinase inhibitor is an RAF inhibitor.
    444. [444] The use according to [443], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    445. [445] The use according to [436], wherein the kinase inhibitor is a VEGFR inhibitor.
    446. [446] The use according to [445], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    447. [447] The use according to [436], wherein the kinase inhibitor is a KIT inhibitor.
    448. [448] The use according to [447], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    449. [449] The use according to [436], wherein the kinase inhibitor is an RET inhibitor.
    450. [450] The use according to [449], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    451. [451] The use according to [436], wherein the kinase inhibitor is a PDGFR inhibitor.
    452. [452] The use according to [451], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    453. [453] The use according to [436], wherein the kinase inhibitor is an FGFR inhibitor.
    454. [454] The use according to [453], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    455. [455] The use according to [436], wherein the kinase inhibitor is an FLT3 inhibitor.
    456. [456] The use according to [455], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    457. [457] The use according to [436], wherein the kinase inhibitor is an ALK inhibitor.
    458. [458] The use according to [457], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    459. [459] The use according to [436], wherein the kinase inhibitor is a CSF-1R inhibitor.
    460. [460] The use according to [459], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    461. [461] The use according to [436], wherein the kinase inhibitor is an EGFR inhibitor.
    462. [462] The use according to [461], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    463. [463] The use according to [436], wherein the kinase inhibitor is an HER2 inhibitor.
    464. [464] The use according to [463], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    465. [465] The use according to any one of [435] to [464], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    466. [466] The use according to [465], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    467. [467] The pharmaceutical composition according to [466], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    468. [468] The pharmaceutical composition according to [466], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    469. [469] The pharmaceutical composition according to [466], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    470. [470] The use according to [466], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    471. [471] The use according to any one of [466] to [470], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    472. [472] The use according to [465], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    473. [473] The use according to [472], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    474. [474] The use according to [473], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    475. [475] The use according to any one of [472] to [474], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    476. [476] The use according to [465], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    477. [477] The use according to [476], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    478. [478] The use according to [477], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    479. [479] The use according to any one of [476] to [478], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    480. [480] The use according to [465], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    481. [481] The use according to [480], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    482. [482] The use according to [481], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    483. [483] The use according to any one of [480] to [482], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    484. [484] The use according to [465], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    485. [485] The use according to [484], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    486. [486] The use according to [485], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    487. [487] The use according to any one of [484] to [486], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    488. [488] The use according to any one of [435] to [487], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    489. [489] The use according to any one of [435] to[488], wherein the use is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    490. [490] The use according to [489], wherein the use is for treating breast cancer.
    491. [491] The use according to [489], wherein the use is for treating colorectal cancer.
    492. [492] The use according to [489], wherein the use is for treating gastric cancer.
    493. [493] The use according to [489], wherein the use is for treating lung cancer.
    494. [494] The use according to [489], wherein the use is for treating pancreatic cancer.
    495. [495] The use according to [489], wherein the use is for treating kidney cancer.
    496. [496] The use according to [489], wherein the use is for treating ovarian cancer.
    497. [497] A pharmaceutical composition wherein an antibody-drug conjugate and a kinase inhibitor are administered in combination, and the antibody-drug conjugate is an antibody-drug conjugate in which a drug-linker represented by the following formula:
      Figure imgb0012
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    498. [498] The pharmaceutical composition according to [497], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    499. [499] The pharmaceutical composition according to [497], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    500. [500] The pharmaceutical composition according to [497], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    501. [501] The pharmaceutical composition according to [497], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    502. [502] The pharmaceutical composition according to [497], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    503. [503] The pharmaceutical composition according to [497], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    504. [504] The pharmaceutical composition according to [497], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    505. [505] The pharmaceutical composition according to [497], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    506. [506] The pharmaceutical composition according to [497], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    507. [507] The pharmaceutical composition according to [497], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    508. [508] The pharmaceutical composition according to [497], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    509. [509] The pharmaceutical composition according to [497], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    510. [510] The pharmaceutical composition according to [497], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    511. [511] The pharmaceutical composition according to [497], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    512. [512] The pharmaceutical composition according to [497], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    513. [513] The pharmaceutical composition according to any one of [497] to [512], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    514. [514] The pharmaceutical composition according to [513], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    515. [515] The pharmaceutical composition according to [514], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    516. [516] The pharmaceutical composition according to [514], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    517. [517] The pharmaceutical composition according to [514], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    518. [518] The pharmaceutical composition according to [514], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    519. [519] The pharmaceutical composition according to any one of [514] to [518], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    520. [520] The pharmaceutical composition according to [513], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    521. [521] The pharmaceutical composition according to [520], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    522. [522] The pharmaceutical composition according to [521], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    523. [523] The pharmaceutical composition according to any one of [520] to [522], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    524. [524] The pharmaceutical composition according to [513], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    525. [525] The pharmaceutical composition according to [524], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    526. [526] The pharmaceutical composition according to [525], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    527. [527] The pharmaceutical composition according to any one of [524] to [526], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    528. [528] The pharmaceutical composition according to [513], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    529. [529] The pharmaceutical composition according to [528], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    530. [530] The pharmaceutical composition according to [529], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    531. [531] The pharmaceutical composition according to any one of [528] to [530], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    532. [532] The pharmaceutical composition according to [513], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    533. [533] The pharmaceutical composition according to [532], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    534. [534] The pharmaceutical composition according to [533], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    535. [535] The pharmaceutical composition according to any one of [532] to [534], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    536. [536] The pharmaceutical composition according to any one of [497] to [535], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    537. [537] The pharmaceutical composition according to any one of [497] to [536], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    538. [538] The pharmaceutical composition according to [537], wherein the pharmaceutical composition is for use in treating breast cancer.
    539. [539] The pharmaceutical composition according to [537], wherein the pharmaceutical composition is for use in treating colorectal cancer.
    540. [540] The pharmaceutical composition according to [537], wherein the pharmaceutical composition is for use in treating gastric cancer.
    541. [541] The pharmaceutical composition according to [537], wherein the pharmaceutical composition is for use in treating lung cancer.
    542. [542] The pharmaceutical composition according to [537], wherein the pharmaceutical composition is for use in treating pancreatic cancer.
    543. [543] The pharmaceutical composition according to [537], wherein the pharmaceutical composition is for use in treating kidney cancer.
    544. [544] The pharmaceutical composition according to [537], wherein the pharmaceutical composition is for use in treating ovarian cancer.
    545. [545] A pharmaceutical composition wherein an antibody-drug conjugate and a kinase inhibitor are administered in combination, and the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0013
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    546. [546] The pharmaceutical composition according to [545], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    547. [547] The pharmaceutical composition according to [545], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    548. [548] The pharmaceutical composition according to [545], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    549. [549] The pharmaceutical composition according to [545], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    550. [550] The pharmaceutical composition according to [545], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    551. [551] The pharmaceutical composition according to [545], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    552. [552] The pharmaceutical composition according to [545], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    553. [553] The pharmaceutical composition according to [545], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    554. [554] The pharmaceutical composition according to [545], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    555. [555] The pharmaceutical composition according to [545], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    556. [556] The pharmaceutical composition according to [545], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    557. [557] The pharmaceutical composition according to [545], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    558. [558] The pharmaceutical composition according to [545], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    559. [559] The pharmaceutical composition according to [545], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    560. [560] The pharmaceutical composition according to [545], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    561. [561] The pharmaceutical composition according to any one of [545] to [560], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    562. [562] The pharmaceutical composition according to [561], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    563. [563] The pharmaceutical composition according to [562], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    564. [564] The pharmaceutical composition according to [562], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    565. [565] The pharmaceutical composition according to [562], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    566. [566] The pharmaceutical composition according to [562], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    567. [567] The pharmaceutical composition according to any one of [562] to [566], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    568. [568] The pharmaceutical composition according to [561], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    569. [569] The pharmaceutical composition according to [568], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    570. [570] The pharmaceutical composition according to [569], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    571. [571] The pharmaceutical composition according to any one of [568] to [570], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    572. [572] The pharmaceutical composition according to [561], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    573. [573] The pharmaceutical composition according to [572], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    574. [574] The pharmaceutical composition according to [573], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    575. [575] The pharmaceutical composition according to any one of [572] to [574], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    576. [576] The pharmaceutical composition according to [561], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    577. [577] The pharmaceutical composition according to [576], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    578. [578] The pharmaceutical composition according to [577], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    579. [579] The pharmaceutical composition according to any one of [576] to [578], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    580. [580] The pharmaceutical composition according to [561], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    581. [581] The pharmaceutical composition according to [580], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    582. [582] The pharmaceutical composition according to [581], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    583. [583] The pharmaceutical composition according to any one of [580] to [582], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    584. [584] The pharmaceutical composition according to any one of [545] to [583], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    585. [585] The pharmaceutical composition according to any one of [545] to [584], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    586. [586] The pharmaceutical composition according to [585], wherein the pharmaceutical composition is for use in treating breast cancer.
    587. [587] The pharmaceutical composition according to [585], wherein the pharmaceutical composition is for use in treating colorectal cancer.
    588. [588] The pharmaceutical composition according to [585], wherein the pharmaceutical composition is for use in treating gastric cancer.
    589. [589] The pharmaceutical composition according to [585], wherein the pharmaceutical composition is for use in treating lung cancer.
    590. [590] The pharmaceutical composition according to [585], wherein the pharmaceutical composition is for use in treating pancreatic cancer.
    591. [591] The pharmaceutical composition according to [585], wherein the pharmaceutical composition is for use in treating kidney cancer.
    592. [592] The pharmaceutical composition according to [585], wherein the pharmaceutical composition is for use in treating ovarian cancer.
    593. [593] A method of treatment, comprising administering an antibody-drug conjugate and a kinase inhibitor in combination to a subject in need of treatment, wherein the antibody-drug conjugate is an antibody-drug conjugate in which a drug-linker represented by the following formula:
      Figure imgb0014
      Figure imgb0015
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    594. [594] The method of treatment according to [593], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    595. [595] The method of treatment according to [593], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    596. [596] The method of treatment according to [593], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    597. [597] The method of treatment according to [593], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    598. [598] The method of treatment according to [593], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    599. [599] The method of treatment according to [593], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    600. [600] The method of treatment according to [593], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    601. [601] The method of treatment according to [593], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    602. [602] The method of treatment according to [593], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    603. [603] The method of treatment according to [593], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    604. [604] The method of treatment according to [593], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    605. [605] The method of treatment according to [593], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    606. [606] The method of treatment according to [593], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    607. [607] The method of treatment according to [593], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    608. [608] The method of treatment according to [593], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    609. [609] The method of treatment according to any one of [593] to [608], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    610. [610] The method of treatment according to [609], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    611. [611] The method of treatment according to [610], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    612. [612] The method of treatment according to [610], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    613. [613] The method of treatment according to [610], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    614. [614] The method of treatment according to [610], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    615. [615] The method of treatment according to any one of [610] to [614], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    616. [616] The method of treatment according to [609], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    617. [617] The method of treatment according to [616], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    618. [618] The method of treatment according to [617], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    619. [619] The method of treatment according to any one of [616] to [618], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    620. [620] The method of treatment according to [609], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    621. [621] The method of treatment according to [620], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    622. [622] The method of treatment according to [621], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    623. [623] The method of treatment according to any one of [620] to [622], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    624. [624] The method of treatment according to [609], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    625. [625] The method of treatment according to [624], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    626. [626] The method of treatment according to [625], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    627. [627] The method of treatment according to any one of [624] to [626], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    628. [628] The method of treatment according to [609], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    629. [629] The method of treatment according to [628], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    630. [630] The method of treatment according to [629], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    631. [631] The method of treatment according to any one of [628] to [630], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    632. [632] The method of treatment according to any one of [593] to [631], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    633. [633] The pharmaceutical composition according to any one of [593] to [632], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    634. [634] The method of treatment according to [633], wherein the method of treatment is for treating breast cancer.
    635. [635] The method of treatment according to [633], wherein the method of treatment is for treating colorectal cancer.
    636. [636] The method of treatment according to [633], wherein the method of treatment is for treating gastric cancer.
    637. [637] The method of treatment according to [633], wherein the method of treatment is for treating lung cancer.
    638. [638] The method of treatment according to [633], wherein the method of treatment is for treating pancreatic cancer.
    639. [639] The method of treatment according to [633], wherein the method of treatment is for treating kidney cancer.
    640. [640] The method of treatment according to [633], wherein the method of treatment is for treating ovarian cancer.
    641. [641] A method of treatment, comprising administering an antibody-drug conjugate and a kinase inhibitor in combination to a subject in need of treatment, wherein the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0016
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    642. [642] The method of treatment according to [641], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    643. [643] The method of treatment according to [641], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    644. [644] The method of treatment according to [641], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    645. [645] The method of treatment according to [641], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    646. [646] The method of treatment according to [641], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    647. [647] The method of treatment according to [641], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    648. [648] The method of treatment according to [641], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    649. [649] The method of treatment according to [641], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    650. [650] The method of treatment according to [641], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    651. [651] The method of treatment according to [641], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    652. [652] The method of treatment according to [641], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    653. [653] The method of treatment according to [641], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    654. [654] The method of treatment according to [641], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    655. [655] The method of treatment according to [641], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    656. [656] The method of treatment according to [641], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    657. [657] The method of treatment according to any one of [641] to [656], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    658. [658] The method of treatment according to [657], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    659. [659] The method of treatment according to [658], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    660. [660] The method of treatment according to [658], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    661. [661] The method of treatment according to [658], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    662. [662] The method of treatment according to [658], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    663. [663] The method of treatment according to any one of [658] to [662], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    664. [664] The method of treatment according to [657], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    665. [665] The method of treatment according to [664], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    666. [666] The method of treatment according to [665], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    667. [667] The method of treatment according to any one of [664] to [666], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    668. [668] The method of treatment according to [657], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    669. [669] The method of treatment according to [668], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    670. [670] The method of treatment according to [669], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    671. [671] The method of treatment according to any one of [668] to [670], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    672. [672] The method of treatment according to [657], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    673. [673] The method of treatment according to [672], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    674. [674] The method of treatment according to [673], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    675. [675] The method of treatment according to any one of [672] to [674], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    676. [676] The method of treatment according to [657], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    677. [677] The method of treatment according to [676], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    678. [678] The method of treatment according to [677], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    679. [679] The method of treatment according to any one of [676] to [678], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    680. [680] The method of treatment according to any one of [641] to [679], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    681. [681] The method of treatment according to any one of [641] to [680], wherein the method of treatment is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    682. [682] The method of treatment according to [681], wherein the method of treatment is for treating breast cancer.
    683. [683] The method of treatment according to [681], wherein the method of treatment is for treating colorectal cancer.
    684. [684] The method of treatment according to [681], wherein the method of treatment is for treating gastric cancer.
    685. [685] The method of treatment according to [681], wherein the method of treatment is for treating lung cancer.
    686. [686] The method of treatment according to [681], wherein the method of treatment is for treating pancreatic cancer.
    687. [687] The method of treatment according to [681], wherein the method of treatment is for treating kidney cancer.
    688. [688] The method of treatment according to [681], wherein the method of treatment is for treating ovarian cancer.
    689. [689] An antibody-drug conjugate for use in treating a disease through being administered in combination with a kinase inhibitor, wherein a drug-linker represented by the following formula:
      Figure imgb0017
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond in the antibody-drug conjugate, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    690. [690] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    691. [691] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    692. [692] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    693. [693] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    694. [694] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    695. [695] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    696. [696] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    697. [697] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    698. [698] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    699. [699] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    700. [700] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    701. [701] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    702. [702] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    703. [703] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    704. [704] The antibody-drug conjugate according to [689], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    705. [705] The antibody-drug conjugate according to any one of [689] to [704], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    706. [706] The antibody-drug conjugate according to [705], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    707. [707] The antibody-drug conjugate according to [706], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    708. [708] The antibody-drug conjugate according to [706], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    709. [709] The antibody-drug conjugate according to [706], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    710. [710] The antibody-drug conjugate according to [706], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    711. [711] The antibody-drug conjugate according to any one of [706] to [710], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    712. [712] The antibody-drug conjugate according to [705], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    713. [713] The antibody-drug conjugate according to [712], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    714. [714] The antibody-drug conjugate according to [713], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    715. [715] The antibody-drug conjugate according to any one of [712] to [714], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    716. [716] The antibody-drug conjugate according to [705], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    717. [717] The antibody-drug conjugate according to [716], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    718. [718] The antibody-drug conjugate according to [717], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    719. [719] The antibody-drug conjugate according to any one of [716] to [718], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    720. [720] The antibody-drug conjugate according to [705], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    721. [721] The antibody-drug conjugate according to [720], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    722. [722] The antibody-drug conjugate according to [721], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    723. [723] The antibody-drug conjugate according to any one of [720] to [722], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    724. [724] The antibody-drug conjugate according to [705], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    725. [725] The antibody-drug conjugate according to [724], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    726. [726] The antibody-drug conjugate according to [725], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    727. [727] The antibody-drug conjugate according to any one of [724] to [726], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    728. [728] The antibody-drug conjugate according to any one of [689] to [727], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    729. [729] The antibody-drug conjugate according to any one of [689] to [728], wherein the antibody-drug conjugate is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    730. [730] The antibody-drug conjugate according to [729], wherein the antibody-drug conjugate is for use in treating breast cancer.
    731. [731] The antibody-drug conjugate according to [729], wherein the antibody-drug conjugate is for use in treating colorectal cancer.
    732. [732] The antibody-drug conjugate according to [729], wherein the antibody-drug conjugate is for use in treating gastric cancer.
    733. [733] The antibody-drug conjugate according to [729], wherein the antibody-drug conjugate is for use in treating lung cancer.
    734. [734] The antibody-drug conjugate according to [729], wherein the antibody-drug conjugate is for use in treating pancreatic cancer.
    735. [735] The antibody-drug conjugate according to [729], wherein the antibody-drug conjugate is for use in treating kidney cancer.
    736. [736] The antibody-drug conjugate according to [729], wherein the antibody-drug conjugate is for use in treating ovarian cancer.
    737. [737] An antibody-drug conjugate for use in treating a disease through being administered in combination with a kinase inhibitor, wherein the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0018
      Figure imgb0019
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    738. [738] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    739. [739] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    740. [740] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    741. [741] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    742. [742] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    743. [743] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    744. [744] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    745. [745] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    746. [746] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    747. [747] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    748. [748] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    749. [749] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    750. [750] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    751. [751] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    752. [752] The antibody-drug conjugate according to [737], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    753. [753] The antibody-drug conjugate according to any one of [737] to [752], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    754. [754] The antibody-drug conjugate according to [753], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    755. [755] The antibody-drug conjugate according to [754], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    756. [756] The antibody-drug conjugate according to [754], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    757. [757] The antibody-drug conjugate according to [754], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    758. [758] The antibody-drug conjugate according to [754], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    759. [759] The antibody-drug conjugate according to any one of [754] to [758], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    760. [760] The antibody-drug conjugate according to [753], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    761. [761] The antibody-drug conjugate according to [760], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    762. [762] The antibody-drug conjugate according to [761], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    763. [763] The antibody-drug conjugate according to any one of [760] to [762], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    764. [764] The antibody-drug conjugate according to [753], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    765. [765] The antibody-drug conjugate according to [764], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    766. [766] The antibody-drug conjugate according to [765], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    767. [767] The antibody-drug conjugate according to any one of [764] to [766], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    768. [768] The antibody-drug conjugate according to [753], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    769. [769] The antibody-drug conjugate according to [768], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    770. [770] The antibody-drug conjugate according to [769], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    771. [771] The antibody-drug conjugate according to any one of [768] to [770], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    772. [772] The antibody-drug conjugate according to [753], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    773. [773] The antibody-drug conjugate according to [772], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    774. [774] The antibody-drug conjugate according to [773], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    775. [775] The antibody-drug conjugate according to any one of [772] to [774], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    776. [776] The antibody-drug conjugate according to any one of [737] to [775], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    777. [777] The antibody-drug conjugate according to any one of [737] to [776], wherein the antibody-drug conjugate is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    778. [778] The antibody-drug conjugate according to [777], wherein the antibody-drug conjugate is for use in treating breast cancer.
    779. [779] The antibody-drug conjugate according to [777], wherein the antibody-drug conjugate is for use in treating colorectal cancer.
    780. [780] The antibody-drug conjugate according to [777], wherein the antibody-drug conjugate is for use in treating gastric cancer.
    781. [781] The antibody-drug conjugate according to [777], wherein the antibody-drug conjugate is for use in treating lung cancer.
    782. [782] The antibody-drug conjugate according to [777], wherein the antibody-drug conjugate is for use in treating pancreatic cancer.
    783. [783] The antibody-drug conjugate according to [777], wherein the antibody-drug conjugate is for use in treating kidney cancer.
    784. [784] The antibody-drug conjugate according to [777], wherein the antibody-drug conjugate is for use in treating ovarian cancer.
    785. [785] Use of an antibody-drug conjugate for the manufacture of a medicament for treating a disease through being administered in combination with a kinase inhibitor, wherein a drug-linker represented by the following formula:
      Figure imgb0020
      wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond in the antibody-drug conjugate, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    786. [786] The use according to [785], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    787. [787] The use according to [785], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    788. [788] The use according to [785], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    789. [789] The use according to [785], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    790. [790] The use according to [785], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    791. [791] The use according to [785], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    792. [792] The use according to [785], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    793. [793] The use according to [785], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    794. [794] The use according to [785], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    795. [795] The use according to [785], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    796. [796] The use according to [785], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    797. [797] The use according to [785], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    798. [798] The use according to [785], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    799. [799] The use according to [785], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    800. [800] The use according to [785], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    801. [801] The use according to any one of [785] to [800], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    802. [802] The use according to [801], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    803. [803] The use according to [802], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    804. [804] The pharmaceutical composition according to [802], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    805. [805] The use according to [802], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 2.
    806. [806] The use according to [802], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    807. [807] The use according to any one of [802] to [806], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    808. [808] The use according to [801], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    809. [809] The use according to [808], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    810. [810] The use according to [809], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    811. [811] The use according to any one of [808] to [810], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    812. [812] The use according to [801], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    813. [813] The use according to [812], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    814. [814] The use according to [813], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    815. [815] The use according to any one of [812] to [814], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    816. [816] The use according to [801], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    817. [817] The use according to [816], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    818. [818] The use according to [817], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    819. [819] The use according to any one of [816] to [818], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    820. [820] The use according to [801], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    821. [821] The use according to [820], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    822. [822] The use according to [821], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    823. [823] The use according to any one of [820] to [822], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    824. [824] The use according to any one of [785] to [823], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    825. [825] The use according to any one of [785] to [824], wherein the use is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    826. [826] The use according to [825], wherein the use is for treating breast cancer.
    827. [827] The use according to [825], wherein the use is for treating colorectal cancer.
    828. [828] The use according to [825], wherein the use is for treating gastric cancer.
    829. [829] The use according to [825], wherein the use is for treating lung cancer.
    830. [830] The use according to [825], wherein the use is for treating pancreatic cancer.
    831. [831] The use according to [825], wherein the use is for treating kidney cancer.
    832. [832] The use according to [825], wherein the use is for treating ovarian cancer.
    833. [833] Use of an antibody-drug conjugate for the manufacture of a medicament for treating a disease through being administered in combination with a kinase inhibitor, wherein the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
      Figure imgb0021
      wherein a drug-linker is conjugated to an antibody via a thioether bond, and n is the average number of units of the drug-linker conjugated per antibody molecule, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    834. [834] The use according to [833], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    835. [835] The use according to [833], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    836. [836] The use according to [833], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    837. [837] The use according to [833], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    838. [838] The use according to [833], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    839. [839] The use according to [833], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    840. [840] The use according to [833], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    841. [841] The use according to [833], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    842. [842] The use according to [833], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    843. [843] The use according to [833], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    844. [844] The use according to [833], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    845. [845] The use according to [833], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    846. [846] The use according to [833], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    847. [847] The use according to [833], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    848. [848] The use according to [833], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    849. [849] The use according to any one of [833] to [848], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
    850. [850] The use according to [849], wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
    851. [851] The use according to [850], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
    852. [852] The use according to [850], wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
    853. [853] The use according to [850], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
    854. [854] The use according to [850], wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
    855. [855] The use according to any one of [850] to [854], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    856. [856] The use according to [849], wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
    857. [857] The use according to [856], wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
    858. [858] The use according to [857], wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    859. [859] The use according to any one of [856] to [858], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    860. [860] The use according to [849], wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
    861. [861] The use according to [860], wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
    862. [862] The use according to [861], wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    863. [863] The use according to any one of [860]to [862], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    864. [864] The use according to [849], wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
    865. [865] The use according to [864], wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
    866. [866] The use according to [865], wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    867. [867] The use according to any one of [864] to [866], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
    868. [868] The use according to [849], wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
    869. [869] The use according to [868], wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
    870. [870] The use according to [869], wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
    871. [871] The use according to any one of [868]to [870], wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
    872. [872] The use according to any one of [833] to [871], wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
    873. [873] The use according to any one of [833] to [872], wherein the use is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    874. [874] The use according to [873], wherein the use is for treating breast cancer.
    875. [875] The use according to [873], wherein the use is for treating colorectal cancer.
    876. [876] The use according to [873], wherein the use is for treating gastric cancer.
    877. [877] The use according to [873], wherein the use is for treating lung cancer.
    878. [878] The use according to [873], wherein the use is for treating pancreatic cancer.
    879. [879] The use according to [873], wherein the use is for treating kidney cancer.
    880. [880] The use according to [873], wherein the use is for treating ovarian cancer.
    881. [881] A pharmaceutical composition wherein an anti-cancer agent and a kinase inhibitor are administered in combination, and the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0022
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    882. [882] The pharmaceutical composition according to [881], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    883. [883] The pharmaceutical composition according to [882], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    884. [884] The pharmaceutical composition according to [883], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    885. [885] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an mTOR inhibitor.
    886. [886] The pharmaceutical composition according to [885], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    887. [887] The pharmaceutical composition according to [882], wherein the kinase inhibitor is a PI3K inhibitor.
    888. [888] The pharmaceutical composition according to [887], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    889. [889] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an RAF inhibitor.
    890. [890] The pharmaceutical composition according to [889], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    891. [891] The pharmaceutical composition according to [882], wherein the kinase inhibitor is a VEGFR inhibitor.
    892. [892] The pharmaceutical composition according to [891], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    893. [893] The pharmaceutical composition according to [882], wherein the kinase inhibitor is a KIT inhibitor.
    894. [894] The pharmaceutical composition according to [893], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    895. [895] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an RET inhibitor.
    896. [896] The pharmaceutical composition according to [895], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    897. [897] The pharmaceutical composition according to [882], wherein the kinase inhibitor is a PDGFR inhibitor.
    898. [898] The pharmaceutical composition according to [897], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    899. [899] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an FGFR inhibitor.
    900. [900] The pharmaceutical composition according to [897], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    901. [901] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an FLT3 inhibitor.
    902. [902] The pharmaceutical composition according to [901], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    903. [903] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an ALK inhibitor.
    904. [904] The pharmaceutical composition according to [903], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    905. [905] The pharmaceutical composition according to [882], wherein the kinase inhibitor is a CSF-1R inhibitor.
    906. [906] The pharmaceutical composition according to [905], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    907. [907] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an EGFR inhibitor.
    908. [908] The pharmaceutical composition according to [907], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    909. [909] The pharmaceutical composition according to [882], wherein the kinase inhibitor is an HER2 inhibitor.
    910. [910] The pharmaceutical composition according to [909], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    911. [911] The pharmaceutical composition according to any one of [881] to [910], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    912. [912] A method of treatment comprising administering an anti-cancer agent and a kinase inhibitor in combination to a subject in need of treatment, wherein the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0023
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    913. [913] The method of treatment according to [912], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    914. [914] The method of treatment according to [913], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    915. [915] The method of treatment according to [914], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    916. [916] The method of treatment according to [913], wherein the kinase inhibitor is an mTOR inhibitor.
    917. [917] The method of treatment according to [916], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    918. [918] The method of treatment according to [913], wherein the kinase inhibitor is a PI3K inhibitor.
    919. [919] The method of treatment according to [918], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    920. [920] The method of treatment according to [913], wherein the kinase inhibitor is an RAF inhibitor.
    921. [921] The method of treatment according to [920], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    922. [922] The method of treatment according to [913], wherein the kinase inhibitor is a VEGFR inhibitor.
    923. [923] The method of treatment according to [922], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    924. [924] The method of treatment according to [913], wherein the kinase inhibitor is a KIT inhibitor.
    925. [925] The method of treatment according to [924], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    926. [926] The method of treatment according to [913], wherein the kinase inhibitor is an RET inhibitor.
    927. [927] The method of treatment according to [926], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    928. [928] The method of treatment according to [913], wherein the kinase inhibitor is a PDGFR inhibitor.
    929. [929] The method of treatment according to [928], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    930. [930] The method of treatment according to [913], wherein the kinase inhibitor is an FGFR inhibitor.
    931. [931] The method of treatment according to [930], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    932. [932] The method of treatment according to [913], wherein the kinase inhibitor is an FLT3 inhibitor.
    933. [933] The method of treatment according to [932], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    934. [934] The method of treatment according to [913], wherein the kinase inhibitor is an ALK inhibitor.
    935. [935] The method of treatment according to [934], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    936. [936] The method of treatment according to [913], wherein the kinase inhibitor is a CSF-1R inhibitor.
    937. [937] The method of treatment according to [936], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    938. [938] The method of treatment according to [913], wherein the kinase inhibitor is an EGFR inhibitor.
    939. [939] The method of treatment according to [938], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    940. [940] The method of treatment according to [913], wherein the kinase inhibitor is an HER2 inhibitor.
    941. [941] The method of treatment according to [940], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    942. [942] The method of treatment according to any one of [912] to [941], wherein the method of treatment is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    943. [943] An anti-cancer agent for use in treating a disease through being administered in combination with a kinase inhibitor, wherein the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0024
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    944. [944] The anti-cancer agent according to [943], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    945. [945] The anti-cancer agent according to [944], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    946. [946] The anti-cancer agent according to [945], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    947. [947] The anti-cancer agent according to [944], wherein the kinase inhibitor is an mTOR inhibitor.
    948. [948] The anti-cancer agent according to [947], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    949. [949] The anti-cancer agent according to [944], wherein the kinase inhibitor is a PI3K inhibitor.
    950. [950] The anti-cancer agent according to [949], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    951. [951] The anti-cancer agent according to [944], wherein the kinase inhibitor is an RAF inhibitor.
    952. [952] The anti-cancer agent according to [951], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    953. [953] The anti-cancer agent according to [944], wherein the kinase inhibitor is a VEGFR inhibitor.
    954. [954] The anti-cancer agent according to [953], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    955. [955] The anti-cancer agent according to [944], wherein the kinase inhibitor is a KIT inhibitor.
    956. [956] The anti-cancer agent according to [955], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    957. [957] The anti-cancer agent according to [944], wherein the kinase inhibitor is an RET inhibitor.
    958. [958] The anti-cancer agent according to [957], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    959. [959] The anti-cancer agent according to [944], wherein the kinase inhibitor is a PDGFR inhibitor.
    960. [960] The anti-cancer agent according to [959], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    961. [961] The anti-cancer agent according to [944], wherein the kinase inhibitor is an FGFR inhibitor.
    962. [962] The anti-cancer agent according to [961], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    963. [963] The anti-cancer agent according to [944], wherein the kinase inhibitor is an FLT3 inhibitor.
    964. [964] The anti-cancer agent according to [963], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    965. [965] The anti-cancer agent according to [944], wherein the kinase inhibitor is an ALK inhibitor.
    966. [966] The anti-cancer agent according to [965], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    967. [967] The anti-cancer agent according to [944], wherein the kinase inhibitor is a CSF-1R inhibitor.
    968. [968] The anti-cancer agent according to [967], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    969. [969] The anti-cancer agent according to [944], wherein the kinase inhibitor is an EGFR inhibitor.
    970. [970] The anti-cancer agent according to [969], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    971. [971] The anti-cancer agent according to [944], wherein the kinase inhibitor is an HER2 inhibitor.
    972. [972] The anti-cancer agent according to [971], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    973. [973] The anti-cancer agent according to any one of [943] to [972], wherein the anti-cancer agent is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    974. [974] Use of an anti-cancer agent for the manufacture of a medicament for treating a disease through being administered in combination with a kinase inhibitor, wherein the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0025
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    975. [975] The use according to [974], wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
    976. [976] The use according to [975], wherein the kinase inhibitor is a CDK4/6 inhibitor.
    977. [977] The use according to [976], wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
    978. [978] The use according to [975], wherein the kinase inhibitor is an mTOR inhibitor.
    979. [979] The use according to [978], wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
    980. [980] The use according to [975], wherein the kinase inhibitor is a PI3K inhibitor.
    981. [981] The use according to [980], wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
    982. [982] The use according to [975], wherein the kinase inhibitor is an RAF inhibitor.
    983. [983] The use according to [982], wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
    984. [984] The use according to [975], wherein the kinase inhibitor is a VEGFR inhibitor.
    985. [985] The use according to [984], wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
    986. [986] The use according to [975], wherein the kinase inhibitor is a KIT inhibitor.
    987. [987] The use according to [986], wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    988. [988] The use according to [975], wherein the kinase inhibitor is an RET inhibitor.
    989. [989] The use according to [988], wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
    990. [990] The use according to [975], wherein the kinase inhibitor is a PDGFR inhibitor.
    991. [991] The use according to [990], wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
    992. [992] The use according to [975], wherein the kinase inhibitor is an FGFR inhibitor.
    993. [993] The use according to [992], wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
    994. [994] The use according to [975], wherein the kinase inhibitor is an FLT3 inhibitor.
    995. [995] The use according to [994], wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
    996. [996] The use according to [975], wherein the kinase inhibitor is an ALK inhibitor.
    997. [997] The use according to [996], wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
    998. [998] The use according to [975], wherein the kinase inhibitor is a CSF-1R inhibitor.
    999. [999] The use according to [998], wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
    1000. [1000] The use according to [975], wherein the kinase inhibitor is an EGFR inhibitor.
    1001. [1001] The use according to [1000], wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    1002. [1002] The use according to [975], wherein the kinase inhibitor is an HER2 inhibitor.
    1003. [1003] The use according to [1002], wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
    1004. [1004] The use according to any one of [974] to [1003], wherein the use is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    1005. [1005] A pharmaceutical composition wherein an anti-cancer agent and a kinase inhibitor are administered in combination, and the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0026
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    1006. [1006] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    1007. [1007] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    1008. [1008] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    1009. [1009] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    1010. [1010] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    1011. [1011] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    1012. [1012] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    1013. [1013] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    1014. [1014] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    1015. [1015] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    1016. [1016] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    1017. [1017] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    1018. [1018] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    1019. [1019] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    1020. [1020] The pharmaceutical composition according to [1005], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    1021. [1021] The pharmaceutical composition according to any one of [1005] to [1020], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    1022. [1022] A method of treatment comprising administering an anti-cancer agent and a kinase inhibitor in combination to a subject in need of treatment, wherein the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0027
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    1023. [1023] The method of treatment according to [1022], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    1024. [1024] The method of treatment according to [1022], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    1025. [1025] The method of treatment according to [1022], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    1026. [1026] The method of treatment according to [1022], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    1027. [1027] The method of treatment according to [1022], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    1028. [1028] The method of treatment according to [1022], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    1029. [1029] The method of treatment according to [1022], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    1030. [1030] The method of treatment according to [1022], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    1031. [1031] The method of treatment according to [1022], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    1032. [1032] The method of treatment according to [1022], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    1033. [1033] The method of treatment according to [1022], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    1034. [1034] The method of treatment according to [1022], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    1035. [1035] The method of treatment according to [1022], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    1036. [1036] The method of treatment according to [1022], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    1037. [1037] The method of treatment according to [1022], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    1038. [1038] The pharmaceutical composition according to any one of [1022] to [1037], wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    1039. [1039] An anti-cancer agent for use in treating a disease through being administered in combination with a kinase inhibitor, wherein the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0028
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    1040. [1040] The anti-cancer agent according to [1039], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    1041. [1041] The anti-cancer agent according to [1039], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    1042. [1042] The anti-cancer agent according to [1039], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    1043. [1043] The anti-cancer agent according to [1039], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    1044. [1044] The anti-cancer agent according to [1039], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    1045. [1045] The anti-cancer agent according to [1039], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    1046. [1046] The anti-cancer agent according to [1039], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    1047. [1047] The anti-cancer agent according to [1039], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    1048. [1048] The anti-cancer agent according to [1039], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    1049. [1049] The anti-cancer agent according to [1039], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    1050. [1050] The anti-cancer agent according to [1039], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    1051. [1051] The anti-cancer agent according to [1039], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    1052. [1052] The anti-cancer agent according to [1039], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    1053. [1053] The anti-cancer agent according to [1039], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    1054. [1054] The anti-cancer agent according to [1039], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    1055. [1055] The anti-cancer agent according to any one of [1039] to [1054], wherein the anti-cancer agent is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, and melanoma.
    1056. [1056] Use of an anti-cancer agent for the manufacture of a medicament for treating a disease through being administered in combination with a kinase inhibitor, wherein the anti-cancer agent releases a drug represented by the following formula:
      Figure imgb0029
      in a tumor, and
      the kinase inhibitor is at least one selected from the group consisting of abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, KRX-0601, everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, rigosertib, ipatasertib, uprosertib, MK-2206, BAY1125976, AZD5363, TAS-117, ONC201, BVD-523, CC-90003, GDC-0994, LY3214996, MK-8353, trametinib, binimetinib, selumetinib, refametinib, pimasertib, cobimetinib, E6201, PD-0325901, RO5126766, GDC-0623, regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, dinaciclib, milciclib, seliciclib, alvocidib, roniciclib, voruciclib, AT7519, PHA-793887, CYC-065, MK-8776, LY2606368, LY2603618, CBP501, GDC-0425, CCT245737, AZD1775, volasertib, alisertib, ilorasertib, ENMD-2076, AMG900, imatinib, dasatinib, bosutinib, nilotinib, ponatinib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ibrutinib, acalabrutinib, tirabrutinib, gilteritinib, quizartinib, midostaurin, brigatinib, crizotinib, ceritinib, alectinib, lorlatinib, ruxolitinib, tofacitinib, baricitinib, pacritinib, capmatinib, tepotinib, pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, entrectinib, GR-389988, gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, poziotinib, tucatinib, and mubritinib, and pharmacologically acceptable salts thereof.
    1057. [1057] The use according to [1056], wherein the kinase inhibitor is abemaciclib or a pharmacologically acceptable salt thereof.
    1058. [1058] The use according to [1056], wherein the kinase inhibitor is palbociclib or a pharmacologically acceptable salt thereof.
    1059. [1059] The use according to [1056], wherein the kinase inhibitor is everolimus or a pharmacologically acceptable salt thereof.
    1060. [1060] The use according to [1056], wherein the kinase inhibitor is taselisib or a pharmacologically acceptable salt thereof.
    1061. [1061] The use according to [1056], wherein the kinase inhibitor is alpelisib or a pharmacologically acceptable salt thereof.
    1062. [1062] The use according to [1056], wherein the kinase inhibitor is regorafenib or a pharmacologically acceptable salt thereof.
    1063. [1063] The use according to [1056], wherein the kinase inhibitor is cabozantinib or a pharmacologically acceptable salt thereof.
    1064. [1064] The use according to [1056], wherein the kinase inhibitor is sunitinib or a pharmacologically acceptable salt thereof.
    1065. [1065] The use according to [1056], wherein the kinase inhibitor is nintedanib or a pharmacologically acceptable salt thereof.
    1066. [1066] The use according to [1056], wherein the kinase inhibitor is brigatinib or a pharmacologically acceptable salt thereof.
    1067. [1067] The use according to [1056], wherein the kinase inhibitor is erlotinib or a pharmacologically acceptable salt thereof.
    1068. [1068] The use according to [1056], wherein the kinase inhibitor is neratinib or a pharmacologically acceptable salt thereof.
    1069. [1069] The use according to [1056], wherein the kinase inhibitor is poziotinib or a pharmacologically acceptable salt thereof.
    1070. [1070] The use according to [1056], wherein the kinase inhibitor is tucatinib or a pharmacologically acceptable salt thereof.
    1071. [1071] The use according to [1056], wherein the kinase inhibitor is mubritinib or a pharmacologically acceptable salt thereof.
    1072. [1072] The use according to any one of [1056] to [1071], wherein the use is for treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
    Advantageous Effects of Invention
  • The present invention can provide a pharmaceutical composition wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination, and/or a method of treatment wherein a specific antibody-drug conjugate and a kinase inhibitor are administered in combination to a subject.
  • Brief Description of Drawings
    • [Figure 1] Figure 1 is a diagram showing the amino acid sequence of a heavy chain of an anti-HER2 antibody (SEQ ID NO: 1).
    • [Figure 2] Figure 2 is a diagram showing the amino acid sequence of a light chain of an anti-HER2 antibody (SEQ ID NO: 2).
    • [Figure 3] Figure 3 is a diagram showing the amino acid sequence of a heavy chain of an anti-HER3 antibody (SEQ ID NO: 3).
    • [Figure 4] Figure 4 is a diagram showing the amino acid sequence of a light chain of an anti-HER3 antibody (SEQ ID NO: 4).
    • [Figure 5] Figure 5 is a diagram showing the amino acid sequence of a heavy chain of an anti-TROP2 antibody (SEQ ID NO: 5).
    • [Figure 6] Figure 6 is a diagram showing the amino acid sequence of a light chain of an anti-TROP2 antibody (SEQ ID NO: 6).
    • [Figure 7] Figure 7 is a diagram showing the amino acid sequence of a heavy chain of an anti-B7-H3 antibody (SEQ ID NO: 7).
    • [Figure 8] Figure 8 is a diagram showing the amino acid sequence of a light chain of an anti-B7-H3 antibody (SEQ ID NO: 8).
    • [Figure 9] Figure 9 is a diagram showing the amino acid sequence of a heavy chain of an anti-CDH6 antibody (SEQ ID NO: 9).
    • [Figure 10] Figure 10 is a diagram showing the amino acid sequence of a light chain of an anti-CDH6 antibody (SEQ ID NO: 10).
    • [Figure 11] Figure 11 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted MDA-MB-453 cells for a single administration group with an antibody-drug conjugate (1) and that with everolimus, and for a combined administration group with the antibody-drug conjugate (1) and everolimus.
    • [Figure 12] Figure 12 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted MDA-MB-453 cells for a single administration group with an antibody-drug conjugate (1) and that with taselisib, and for a combined administration group with the antibody-drug conjugate (1) and taselisib.
    • [Figure 13] Figure 13 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted MDA-MB-453 cells for a single administration group with an antibody-drug conjugate (1) and that with abemaciclib, and for a combined administration group with the antibody-drug conjugate (1) and abemaciclib.
    • [Figure 14] Figure 14 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted JIMT-1 cells for a single administration group with an antibody-drug conjugate (1) and that with everolimus, and for a combined administration group with the antibody-drug conjugate (1) and everolimus.
    • [Figure 15] Figure 15 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted COLO201 cells for a single administration group with an antibody-drug conjugate (1) and that with regorafenib, and for a combined administration group with the antibody-drug conjugate (1) and regorafenib.
    • [Figure 16] Figure 16 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted KPL-4 cells for a single administration group with an antibody-drug conjugate (1) and that with tucatinib, and for a combined administration group with the antibody-drug conjugate (1) and tucatinib.
    • [Figure 17] Figure 17 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted MDA-MB-453 cells for a single administration group with an antibody-drug conjugate (1) and that with tucatinib, and for a combined administration group with the antibody-drug conjugate (1) and tucatinib.
    • [Figure 18] Figure 18 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted NCI-N87 cells for a single administration group with an antibody-drug conjugate (1) and that with tucatinib, and for a combined administration group with the antibody-drug conjugate (1) and tucatinib.
    • [Figure 19] Figure 19 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted KPL-4 cells for a single administration group with an antibody-drug conjugate (2) and that with neratinib, and for a combined administration group with the antibody-drug conjugate (2) and neratinib.
    • [Figure 20] Figure 20 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted 786-O cells for a single administration group with an antibody-drug conjugate (3) and that with cabozantinib, and for a combined administration group with the antibody-drug conjugate (3) and cabozantinib.
    • [Figure 21] Figure 21 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted MDA-MB-453 cells for a single administration group with an antibody-drug conjugate (2) and that with palbociclib, and for a combined administration group with the antibody-drug conjugate (2) and palbociclib.
    • [Figure 22] Figure 22 is a diagram showing tumor growth inhibitory effects on mice with subcutaneously transplanted MDA-MB-453 cells for a single administration group with an antibody-drug conjugate (2) and that with alpelisib, and for a combined administration group with the antibody-drug conjugate (2) and alpelisib.
    Description of Embodiments
  • Hereinafter, preferred modes for carrying out the present invention are described. The embodiments described below are given merely for illustrating one example of a typical embodiment of the present invention and are not intended to limit the scope of the present invention.
  • 1. Antibody-drug conjugate
  • The antibody-drug conjugate used in the present invention is an antibody-drug conjugate in which a drug-linker represented by the following formula:
    Figure imgb0030
    • wherein A represents the connecting position to an antibody,
    • is conjugated to the antibody via a thioether bond.
  • In the present invention, the partial structure consisting of a linker and a drug in the antibody-drug conjugate is referred to as a "drug-linker". The drug-linker is connected to a thiol group (in other words, the sulfur atom of a cysteine residue) formed at an interchain disulfide bond site (two sites between heavy chains, and two sites between a heavy chain and a light chain) in the antibody.
  • The drug-linker of the present invention includes exatecan (IUPAC name: (1S,9S)-1-amino-9-ethyl-5-fluoro-1,2,3,9,12,15-hexahydro-9-hydroxy-4-methyl-10H,13H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-10,13-dione, (also expressed as chemical name: (1S,9S)-1-amino-9-ethyl-5-fluoro-2,3-dihydro-9-hydroxy-4-methyl-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-10,13(9H,15H)-dione)), which is a topoisomerase I inhibitor, as a component. Exatecan is a camptothecin derivative having an antitumor effect, represented by the following formula:
    Figure imgb0031
  • The antibody-drug conjugate used in the present invention can be also represented by the following formula:
    Figure imgb0032
    Figure imgb0033
    wherein, the drug-linker is conjugated to an antibody via a thioether bond. The meaning of n is the same as that of what is called the average number of conjugated drug molecules (DAR; Drug-to-Antibody Ratio), and indicates the average number of units of the drug-linker conjugated per antibody molecule.
  • After migrating into cancer cells, the antibody-drug conjugate used in the present invention is cleaved at the linker portion to release the compound represented by the following formula.
    Figure imgb0034
  • The aforementioned compound is inferred to be the original source of the antitumor activity of the antibody-drug conjugate used in the present invention, and has been confirmed to have a topoisomerase I inhibitory effect (Ogitani Y. et al., Clinical Cancer Research, 2016, ) .
  • The antibody-drug conjugate used in the present invention is known to have a bystander effect (Ogitani Y. et al., Cancer Science (2016) 107, 1039-1046).
  • The bystander effect is exerted through a process such that the antibody-drug conjugate used in the present invention is internalized in cancer cells expressing a target and the aforementioned compound is released then exerts an antitumor effect also on cancer cells which are present therearound and not expressing the target.
  • This bystander effect is exerted as an excellent antitumor effect even when the antibody-drug conjugate is used in combination with a kinase inhibitor according to the present invention.
  • 2. Antibody in antibody-drug conjugate
  • The antibody in the antibody-drug conjugate used in the present invention may be derived from any species, and is preferably an antibody derived from a human, a rat, a mouse, or a rabbit. In cases when the antibody is derived from species other than human species, it is preferably chimerized or humanized using a well known technique. The antibody of the present invention may be a polyclonal antibody or a monoclonal antibody and is preferably a monoclonal antibody.
  • The antibody in the antibody-drug conjugate used in the present invention is an antibody preferably having a characteristic of being capable of targeting cancer cells, and is preferably an antibody possessing, for example, a property of recognizing a cancer cell, a property of binding to a cancer cell, a property of internalizing in a cancer cell, and/or cytocidal activity against cancer cells.
  • The binding activity of the antibody against cancer cells can be confirmed using flow cytometry. The internalization of the antibody into cancer cells can be confirmed using (1) an assay of visualizing an antibody incorporated in cells under a fluorescence microscope using a secondary antibody (fluorescently labeled) binding to the therapeutic antibody (Cell Death and Differentiation (2008) 15, 751-761), (2) an assay of measuring a fluorescence intensity incorporated in cells using a secondary antibody (fluorescently labeled) binding to the therapeutic antibody (Molecular Biology of the Cell, Vol. 15, 5268-5282, December 2004), or (3) a Mab-ZAP assay using an immunotoxin binding to the therapeutic antibody wherein the toxin is released upon incorporation into cells to inhibit cell growth (Bio Techniques 28: 162-165, January 2000). As the immunotoxin, a recombinant complex protein of a diphtheria toxin catalytic domain and protein G may be used.
  • The antitumor activity of the antibody can be confirmed in vitro by determining inhibitory activity against cell growth. For example, a cancer cell line overexpressing a target protein for the antibody is cultured, and the antibody is added at varying concentrations into the culture system to determine inhibitory activity against focus formation, colony formation, and spheroid growth. The antitumor activity can be confirmed in vivo, for example, by administering the antibody to a nude mouse with a transplanted cancer cell line highly expressing the target protein, and determining change in the cancer cell.
  • Since the compound conjugated in the antibody-drug conjugate exerts an antitumor effect, it is preferred but not essential that the antibody itself should have an antitumor effect. For the purpose of specifically and selectively exerting the cytotoxic activity of the antitumor compound against cancer cells, it is important and also preferred that the antibody should have the property of internalizing to migrate into cancer cells.
  • The antibody in the antibody-drug conjugate used in the present invention can be obtained by a procedure known in the art. For example, the antibody of the present invention can be obtained using a method usually carried out in the art, which involves immunizing animals with an antigenic polypeptide and collecting and purifying antibodies produced in vivo. The origin of the antigen is not limited to humans, and the animals may be immunized with an antigen derived from a non-human animal such as a mouse, a rat and the like. In this case, the cross-reactivity of antibodies binding to the obtained heterologous antigen with human antigens can be tested to screen for an antibody applicable to a human disease.
  • Alternatively, antibody-producing cells which produce antibodies against the antigen are fused with myeloma cells according to a method known in the art (e.g., Kohler and Milstein, Nature (1975) 256, p. 495-497; and Kennet, R. ed., Monoclonal Antibodies, p. 365-367, Plenum Press, N.Y. (1980)) to establish hybridomas, from which monoclonal antibodies can in turn be obtained.
  • The antigen can be obtained by genetically engineering host cells to produce a gene encoding the antigenic protein. Specifically, vectors that permit expression of the antigen gene are prepared and transferred to host cells so that the gene is expressed. The antigen thus expressed can be purified. The antibody can also be obtained by a method of immunizing animals with the above-described genetically engineered antigenexpressing cells or a cell line expressing the antigen.
  • The antibody in the antibody-drug conjugate used in the present invention is preferably a recombinant antibody obtained by artificial modification for the purpose of decreasing heterologous antigenicity to humans such as a chimeric antibody or a humanized antibody, or is preferably an antibody having only the gene sequence of an antibody derived from a human, that is, a human antibody. These antibodies can be produced using a known method.
  • As the chimeric antibody, an antibody in which antibody variable and constant regions are derived from different species, for example, a chimeric antibody in which a mouse- or rat-derived antibody variable region is connected to a human-derived antibody constant region can be exemplified (Proc. Natl. Acad. Sci. USA, 81, 6851-6855, (1984)).
  • As the humanized antibody, an antibody obtained by integrating only the complementarity determining region (CDR) of a heterologous antibody into a human-derived antibody (Nature (1986) 321, pp. 522-525), and an antibody obtained by grafting a part of the amino acid residues of the framework of a heterologous antibody as well as the CDR sequence of the heterologous antibody to a human antibody by a CDR-grafting method (International Publication No. WO 90/07861 ), and an antibody humanized using a gene conversion mutagenesis strategy ( U.S. Patent No. 5821337 ) can be exemplified.
  • As the human antibody, an antibody generated by using a human antibody-producing mouse having a human chromosome fragment including genes of a heavy chain and light chain of a human antibody (see Tomizuka, K. et al., Nature Genetics (1997) 16, p.133-143; Kuroiwa, Y. et. al., Nucl. Acids Res. (1998) 26, p.3447-3448; Yoshida, H. et. al., Animal Cell Technology:Basic and Applied Aspects vol.10, p.69-73 (Kitagawa, Y., Matsuda, T. and Iijima, S. eds.), Kluwer Academic Publishers, 1999; Tomizuka, K. et. al., Proc. Natl. Acad. Sci. USA (2000) 97, p.722-727, etc.) can be exemplified. As an alternative, an antibody obtained by phage display, the antibody being selected from a human antibody library (see Wormstone, I. M. et. al, Investigative Ophthalmology & Visual Science. (2002)43 (7), p.2301-2308; Carmen, S. et. al., Briefings in Functional Genomics and Proteomics (2002), 1(2), p.189-203; Siriwardena, D. et. al., Ophthalmology (2002) 109(3), p.427-431, etc.) can be exemplified.
  • In the antibody in the antibody-drug conjugate used in present invention, modified variants of the antibody are also included. The modified variant refers to a variant obtained by subjecting the antibody according to the present invention to chemical or biological modification. Examples of the chemically modified variant include variants including a linkage of a chemical moiety to an amino acid skeleton, variants including a linkage of a chemical moiety to an N-linked or O-linked carbohydrate chain, etc. Examples of the biologically modified variant include variants obtained by post-translational modification (such as N-linked or O-linked glycosylation, N- or C-terminal processing, deamidation, isomerization of aspartic acid, or oxidation of methionine), and variants in which a methionine residue has been added to the N terminus by being expressed in a prokaryotic host cell. Further, an antibody labeled so as to enable the detection or isolation of the antibody or an antigen according to the present invention, for example, an enzyme-labeled antibody, a fluorescence-labeled antibody, and an affinity-labeled antibody are also included in the meaning of the modified variant. Such a modified variant of the antibody according to the present invention is useful for improving the stability and blood retention of the antibody, reducing the antigenicity thereof, detecting or isolating an antibody or an antigen, and so on.
  • Further, by regulating the modification of a glycan which is linked to the antibody according to the present invention (glycosylation, defucosylation, etc.), it is possible to enhance antibody-dependent cellular cytotoxic activity. As the technique for regulating the modification of a glycan of antibodies, International Publication No. WO 99/54342 , International Publication No. WO 00/61739 , International Publication No. WO 02/31140 , International Publication No. WO 2007/133855 , International Publication No. WO 2013/120066 , etc. are known. However, the technique is not limited thereto. In the antibody according to the present invention, antibodies in which the modification of a glycan is regulated are also included.
  • It is known that a lysine residue at the carboxyl terminus of the heavy chain of an antibody produced in a cultured mammalian cell is deleted (Journal of Chromatography A, 705: 129-134 (1995)), and it is also known that two amino acid residues (glycine and lysine) at the carboxyl terminus of the heavy chain of an antibody produced in a cultured mammalian cell are deleted and a proline residue newly located at the carboxyl terminus is amidated (Analytical Biochemistry, 360: 75-83 (2007)). However, such deletion and modification of the heavy chain sequence do not affect the antigen-binding affinity and the effector function (the activation of complement, antibody-dependent cellular cytotoxicity, etc.) of the antibody. Therefore, in the antibody according to the present invention, antibodies subjected to such modification and functional fragments of the antibody are also included, and deletion variants in which one or two amino acids have been deleted at the carboxyl terminus of the heavy chain, variants obtained by amidation of deletion variants (for example, a heavy chain in which the carboxyl terminal proline residue has been amidated), and the like are also included. The type of deletion variant having a deletion at the carboxyl terminus of the heavy chain of the antibody according to the present invention is not limited to the above variants as long as the antigen-binding affinity and the effector function are conserved. The two heavy chains constituting the antibody according to the present invention may be of one type selected from the group consisting of a full-length heavy chain and the above-described deletion variant, or may be of two types in combination selected therefrom. The ratio of the amount of each deletion variant can be affected by the type of cultured mammalian cells which produce the antibody according to the present invention and the culture conditions. However, an antibody in which one amino acid residue at the carboxyl terminus has been deleted in both of the two heavy chains in the antibody according to the present invention can be preferably exemplified.
  • As isotypes of the antibody according to the present invention, for example, IgG (IgG1, IgG2, IgG3, IgG4) can be exemplified. Preferably, IgG1 or IgG2 can be exemplified.
  • Examples of antibodies in the antibody-drug conjugate used in the present invention include, but are not limited to, an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, an anti-CDH6 antibody, an anti-CD3 antibody, an anti-CD30 antibody, an anti-CD33 antibody, an anti-CD37 antibody, an anti-CD56 antibody, an anti-CD98 antibody, an anti-DR5 antibody, an anti-EGFR antibody, an anti-EPHA2 antibody, an anti-FGFR2 antibody, an anti-FGFR4 antibody, an anti-FOLR1 antibody, an anti-VEGF antibody, an anti-CD20 antibody, an anti-CD22 antibody, an anti-CD70 antibody, an anti-PSMA antibody, an anti-CEA antibody, an anti-Mesothelin antibody, an anti-A33 antibody, an anti-CanAg antibody, an anti-Cripto antibody, an anti-G250 antibody, an anti-MUC1 antibody, an anti-GPNMB antibody, an anti-Integrin antibody, an anti-Tenascin-C antibody, and an anti-SLC44A4 antibody. Further an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, and an anti-CDH6 antibody can be preferably exemplified.
  • In the present invention, the term "anti-HER2 antibody" refers to an antibody which binds specifically to HER2 (Human Epidermal Growth Factor Receptor Type 2; ErbB-2), and preferably has an activity of internalization in HER2-expressing cells by binding to HER2.
  • Examples of the anti-HER2 antibody include trastuzumab ( U.S. Patent No. 5821337 ) and pertuzumab (International Publication No. WO 01/00245 ). Preferably, trastuzumab can be exemplified.
  • In the present invention, the term "anti-HER3 antibody" refers to an antibody which binds specifically to HER3 (Human Epidermal Growth Factor Receptor Type 3; ErbB-3), and preferably has an activity of internalization in HER3-expressing cells by binding to HER3.
  • Examples of the anti-HER3 antibody include patritumab (U3-1287), U1-59 (International Publication No. WO 2007/077028 ), MM-121 (seribantumab), an anti-ERBB3 antibody described in International Publication No. WO 2008/100624 , RG-7116 (lumretuzumab), and LJM-716 (elgemtumab). Preferably, patritumab and U1-59 can be exemplified.
  • In the present invention, the term "anti-TROP2 antibody" refers to an antibody which binds specifically to TROP2 (TACSTD2: Tumor-associated calcium signal transducer 2; EGP-1), and preferably has an activity of internalization in TROP2-expressing cells by binding to TROP2.
  • Examples of the anti-TROP2 antibody include hTINA1-H1L1 (International Publication No. WO 2015/098099 ).
  • In the present invention, the term "anti-B7-H3 antibody" refers to an antibody which binds specifically to B7-H3 (B cell antigen #7 homolog 3; PD-L3; CD276), and preferably has an activity of internalization in B7-H3-expressing cells by binding to B7-H3.
  • Examples of the anti-B7-H3 antibody include M30-H1-L4 (International Publication No. WO 2014/057687 ).
  • In the present invention, the term "anti-CDH6 antibody" refers to an antibody which binds specifically to CDH6 (Cadherin-6), and preferably has an activity of internalization in CDH6-expressing cells by binding to CDH6.
  • Examples of the anti-CDH6 antibody include H01L02 (International Publication No. WO 2018/212136 ).
  • 3. Production of antibody-drug conjugate
  • A drug-linker intermediate for use in production of the antibody-drug conjugate according to the present invention is represented by the following formula.
    Figure imgb0035
  • The drug-linker intermediate can be expressed as the chemical name N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]glycylglycyl-L-phenylalanyl-N-[(2-{[(1S,9S)-9-ethyl-5-fluoro-9-hydroxy-4-methyl-10,13-dioxo-2,3,9,10,13,15-hexahydro-1H,12H-benzo[de]pyrano[3',4':6,7]indolizino[1,2-b]quinolin-1-yl]amino}-2-oxoethoxy)methyl]glycinamide, and can be produced with reference to descriptions in International Publication No. WO 2014/057687 , International Publication No. WO 2015/098099 , International Publication No. WO 2015/115091 , International Publication No. WO 2015/155998 , International Publication No. WO 2019/044947 , and so on.
  • The antibody-drug conjugate used in the present invention can be produced by reacting the above-described drug-linker intermediate and an antibody having a thiol group (alternatively referred to as a sulfhydryl group).
  • The antibody having a sulfhydryl group can be obtained by a method well known in the art (Hermanson, G. T, Bioconjugate Techniques, pp. 56-136, pp. 456-493, Academic Press (1996)). For example, by using 0.3 to 3 molar equivalents of a reducing agent such as tris(2-carboxyethyl)phosphine hydrochloride (TCEP) per interchain disulfide within the antibody and reacting with the antibody in a buffer solution containing a chelating agent such as ethylenediamine tetraacetic acid (EDTA), an antibody having a sulfhydryl group with partially or completely reduced interchain disulfides within the antibody can be obtained.
  • Further, by using 2 to 20 molar equivalents of the drug-linker intermediate per the antibody having a sulfhydryl group, an antibody-drug conjugate in which 2 to 8 drug molecules are conjugated per antibody molecule can be produced.
  • The average number of conjugated drug molecules per antibody molecule of the antibody-drug conjugate produced can be determined, for example, by a method of calculation based on measurement of UV absorbance for the antibody-drug conjugate and the conjugation precursor thereof at two wavelengths of 280 nm and 370 nm (UV method), or a method of calculation based on quantification through HPLC measurement for fragments obtained by treating the antibody-drug conjugate with a reducing agent (HPLC method).
  • Conjugation between the antibody and the drug-linker intermediate and calculation of the average number of conjugated drug molecules per antibody molecule of the antibody-drug conjugate can be performed with reference to descriptions in International Publication No. WO 2014/057687 , International Publication No. WO 2015/098099 , International Publication No. WO 2015/115091 , International Publication No. WO 2015/155998 , International Publication No. WO 2017/002776 , International Publication No. WO 2018/212136 , and so on.
  • In the present invention, the term "anti-HER2 antibody-drug conjugate" refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate according to the present invention is an anti-HER2 antibody.
  • The anti-HER2 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2;
    • more preferably an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2; and
    • even more preferably an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2, or an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
  • The average number of units of the drug-linker conjugated per antibody molecule in the anti-HER2 antibody-drug conjugate is preferably 2 to 8, more preferably 3 to 8, even more preferably 7 to 8, even more preferably 7.5 to 8, and even more preferably about 8.
  • The anti-HER2 antibody-drug conjugate used in the present invention can be produced with reference to descriptions in International Publication No. WO 2015/115091 and so on.
  • In the present invention, the term "anti-HER3 antibody-drug conjugate" refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate according to the present invention is an anti-HER3 antibody.
  • The anti-HER3 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 35 of SEQ ID NO: 3, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 50 to 65 of SEQ ID NO: 3, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 98 to 106 of SEQ ID NO: 3, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 24 to 39 of SEQ ID NO: 4, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 56 to 62 of SEQ ID NO: 4, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 95 to 103 of SEQ ID NO: 4;
    • more preferably an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 117 of SEQ ID NO: 3 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 113 of SEQ ID NO: 4; and
    • even more preferably an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 4, or a variant of the antibody in which the lysine residue at the carboxyl terminus of the heavy chain is deleted.
  • The average number of units of the drug-linker conjugated per antibody molecule in the anti-HER3 antibody-drug conjugate is preferably 2 to 8, more preferably 3 to 8, even more preferably 7 to 8, even more preferably 7.5 to 8, and even more preferably about 8.
  • The anti-HER3 antibody-drug conjugate can be produced with reference to descriptions in International Publication No. WO 2015/155998 and so on.
  • In the present invention, the term "anti-TROP2 antibody-drug conjugate" refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
  • The anti-TROP2 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 50 to 54 of SEQ ID NO: 5, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 69 to 85 of SEQ ID NO: 5, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 118 to 129 of SEQ ID NO: 5, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 44 to 54 of SEQ ID NO: 6, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 70 to 76 of SEQ ID NO: 6, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 109 to 117 of SEQ ID NO: 6;
    • more preferably an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 20 to 140 of SEQ ID NO: 5 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 21 to 129 of SEQ ID NO: 6; and
    • even more preferably an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6, or a variant of the antibody in which the lysine residue at the carboxyl terminus of the heavy chain is deleted.
  • The average number of units of the drug-linker conjugated per antibody molecule in the anti-TROP2 antibody-drug conjugate used in the present invention is preferably 2 to 8, more preferably 3 to 5, even more preferably 3.5 to 4.5, and even more preferably about 4.
  • The anti-TROP2 antibody-drug conjugate can be produced with reference to descriptions in International Publication No. WO 2015/098099 , International Publication No. WO 2017/002776 , and so on.
  • In the present invention, the term "anti-B7-H3 antibody-drug conjugate" refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate according to the present invention is an anti-B7-H3 antibody.
  • The anti-B7-H3 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 50 to 54 of SEQ ID NO: 7, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 69 to 85 of SEQ ID NO: 7, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 118 to 130 of SEQ ID NO: 7, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 44 to 53 of SEQ ID NO: 8, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 69 to 75 of SEQ ID NO: 8, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 108 to 116 of SEQ ID NO: 8;
    • more preferably an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 20 to 141 of SEQ ID NO: 7 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 21 to 128 of SEQ ID NO: 8; and
    • even more preferably an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8, or a variant of the antibody in which the lysine residue at the carboxyl terminus of the heavy chain is deleted.
  • The average number of units of the drug-linker conjugated per antibody molecule in the anti-B7-H3 antibody-drug conjugate is preferably 2 to 8, more preferably 3 to 5, even more preferably 3.5 to 4.5, and even more preferably about 4.
  • The anti-B7-H3 antibody-drug conjugate used in the present invention can be produced with reference to descriptions in International Publication No. WO 2014/057687 , International Publication No. WO 2017/002776 , and so on.
  • In the present invention, the term "anti-CDH6 antibody-drug conjugate" refers to an antibody-drug conjugate such that the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
  • The anti-CDH6 antibody is preferably an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 45 to 54 of SEQ ID NO: 9, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 69 to 78 of SEQ ID NO: 9, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 118 to 130 of SEQ ID NO: 9, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 44 to 54 of SEQ ID NO: 10, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 70 to 76 of SEQ ID NO: 10, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 109 to 116 of SEQ ID NO: 10;
    • more preferably an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 20 to 141 of SEQ ID NO: 9 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 21 to 128 of SEQ ID NO: 10; and
    • even more preferably an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10, or a variant of the antibody in which the lysine residue at the carboxyl terminus of the heavy chain is deleted.
  • The average number of units of the drug-linker conjugated per antibody molecule in the anti-CDH6 antibody-drug conjugate used in the present invention is preferably 2 to 8, more preferably 3 to 8, even more preferably 7.5 to 8, and even more preferably about 8.
  • The anti-CDH6 antibody-drug conjugate used in the present invention can be produced with reference to descriptions in International Publication No. WO 2018/212136 and so on.
  • 4. Kinase inhibitor
  • In the present invention, the term "kinase inhibitor" refers to an agent that inhibits a kinase involved in cancer cell growth and tumor angiogenesis. As the kinase inhibitor in the present invention, at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor can be exemplified.
  • The term "pharmacologically acceptable salt" of the kinase inhibitor used in the present invention may be either an acid addition salt or a base addition salt. Examples of the acid addition salt can include lower alkanesulfonates such as camsylate (camphorsulfonate), mesylate (methanesulfonate), trifluoromethanesulfonate, and ethanesulfonate; arylsulfonates such as tosylate (p-toluenesulfonate) and benzenesulfonate; inorganic acid salts such as phosphate, nitrate, perchlorate, and sulfate; hydrogen halide salts such as hydrochloride, hydrobromide, hydroiodide, and hydrofluoride; organic acid salts such as acetate, malate, fumarate, succinate, citrate, tartrate, oxalate, and maleate; and amino acid salts such as ornithinate, glutamate, and aspartate. Examples of the base addition salt can include alkali metal salts such as sodium salt, potassium salt, and lithium salt; alkali earth metal salts such as calcium salt and magnesium salt; inorganic salts such as ammonium salt; organic amine salts such as dibenzylamine salt, morpholine salt, phenylglycine alkyl ester salt, ethylenediamine salt, N-methylglucamine salt, diethylamine salt, triethylamine salt, cyclohexylamine salt, dicyclohexylamine salt, N,N'-dibenzylethylenediamine salt, diethanolamine salt, N-benzyl-N-(2-phenylethoxy)amine salt, piperazine salt, tetramethylammonium salt, and tris(hydroxymethyl)aminomethane salt; and amino acid salts such as alginate.
  • The kinase inhibitor and pharmacologically acceptable salts thereof used in the present invention may each exist as a solvate, and solvates of them are also included in the meaning of the kinase inhibitor and pharmacologically acceptable salts thereof used in the present invention.
  • In the present invention, the term "CDK4/6 inhibitor" refers to an agent that inhibits cyclin dependent kinase 4 (CDK4) and inhibits cyclin dependent kinase 6 (CDK6). The CDK4/6 inhibitor in the present invention may have an effect of inhibiting a kinase other than CDK4/6. The CDK4/6 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include abemaciclib ( U.S. Patent No. 7855211 ), palbociclib ( U.S. Patent No. 6936612 ), ribociclib ( U.S. Patent No. 9193732 ), trilaciclib (International Publication No. WO 2012/061156 ), G1T38 (Oncotarget 2017, 8(26): 42343-42358), PF-06873600 (International Publication No. WO 2018/033815 ), TP-1287 (International Publication No. WO 2016/187316 ), FN-1501 (J Med Chem 2018, 61(4): 1499-1518), and KRX-0601 (International Publication No. WO 1989/007105 ), and pharmacologically acceptable salts thereof.
  • PF-06873600 and FN-1501 and pharmacologically acceptable salts thereof are each also known as a CDK2 inhibitor. TP-1287 and pharmacologically acceptable salts thereof are each also known as a CDK1 inhibitor and a CDK2 inhibitor. KRX-0601 and pharmacologically acceptable salts thereof are each also known as a PI3K inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, and a CHK1 inhibitor.
  • In the present invention, the term "mTOR inhibitor" refers to an agent that inhibits mTOR (Mammalian target of Rapamycin), one of the serine/threonine kinases. The mTOR inhibitor in the present invention may have an effect of inhibiting a kinase other than mTOR. The mTOR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include everolimus ( U.S. Patent No. 5665772 ), sirolimus ( U.S. Patent No. 5989591 ), temsirolimus ( U.S. Patent No. 5362718 ), TAK-228 (Cancer Chemother Pharmacol 2017, 80(2): 261-273), CC-223 (J Med Chem 2015, 58(13): 5323-5333), AZD8055 (Cancer Res 2010, 70(1): 288-98), dactolisib (Invest New Drugs 2015, 33(2): 463-71), apitolisib (International Publication No. WO 2008/070740 ), gedatolisib (International Publication No. WO 2009/143313 ), LY3023414 (Mol Cancer Ther 2016, 15(10): 2344-2356), PF-04691502 (Mol Cancer Ther 2011, 10(11): 2189-99), NVP-BGT226 (Ann Oncol 2012, 23(9): 2399-408), and PQR309 (J Med Chem 2017, 60(17): 7524-7538), and pharmacologically acceptable salts thereof.
  • Dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, and PQR309, and pharmacologically acceptable salts thereof are each also known as a PI3K inhibitor.
  • In the present invention, the term "PI3K inhibitor" refers to an agent that inhibits PI3K (Phosphoinositide 3-kinase), an enzyme that phosphorylates the hydroxyl group at position 3 of the inositol ring of inositol phospholipid. The PI3K inhibitor in the present invention may have an effect of inhibiting a kinase other than PI3K. The PI3K inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include taselisib (International Publication No. WO 2014/140073 ), alpelisib (International Publication No. WO 2010/029082 ), TAK-117 (Org Process Res Dev 2017, 21(4): 669-673), GSK2636771 (Clin Cancer Res 2017, 23(19): 5981-92), AZD8186 (J Med Chem 2015, 58(2): 943-962), IPI-549 (ACS Med Chem Lett 2016, 7(9): 862-867), idelalisib ( U.S. Patent No. 9469643 ), duvelisib ( U.S. Patent No. 8193182 ), AMG319 (J Med Chem 2015, 58(1): 480-511), buparlisib (International Publication No. WO 2007/084786 ), pictilisib (International Publication No. WO 2007/129161 ), pilaralisib (International Publication No. WO 2007/044729 ), copanlisib ( U.S. Patent No. 7511041 ), sonolisib (International Publication No. WO 2003/024183 ), CH5132799 (Bioorg Med Chem Lett 2011, 21(6): 1767-1772), ZSTK474 (J Natl Cancer Inst 2006, 98(8): 545-56), GDC-0077 (International Publication No. WO 2017/001645 ), dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, and rigosertib (International Publication No. WO 2006/010152 ), and pharmacologically acceptable salts thereof.
  • Rigosertib and pharmacologically acceptable salts thereof are each also known as a CDK1 inhibitor and a PLK1 inhibitor.
  • The pharmacologically acceptable salt of copanlisib is preferably a hydrochloride (copanlisib hydrochloride). The pharmacologically acceptable salt of rigosertib is preferably a sodium salt (rigosertib sodium).
  • In the present invention, the term "AKT inhibitor" refers to an agent that inhibits AKT (also called protein kinase B), a serine/threonine kinase. The AKT inhibitor in the present invention may have an effect of inhibiting a kinase other than AKT. The AKT inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ipatasertib (International Publication No. WO 2008/006040 ), uprosertib (International Publication No. WO 2008/098104 ), MK-2206 (Mol Cancer Ther 2010, 9(7): 1956-67), BAY1125976 (Int J Cancer 2017, 140(2): 449-454), AZD5363 (Mol Cancer Ther 2012, 11(4): 873-87), and TAS-117 (Cancer Res 2014, 74(16): 4458-69), and pharmacologically acceptable salts thereof.
  • The pharmacologically acceptable salt of ipatasertib is preferably a hydrochloride (ipatasertib hydrochloride).
  • In the present invention, the term "ERK inhibitor" refers to an agent that inhibits extracellular signal-regulated kinase (ERK). The ERK inhibitor in the present invention may have an effect of inhibiting a kinase other than ERK. The ERK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ONC201 (Oncotarget 2014, 5(24): 12728-37), BVD-523 (Mol Cancer Ther 2017, 16(11): 2351-2363), CC-90003 (International Publication No. WO 2014/124230 ), GDC-0994 (J Med Chem 2016, 59(12): 5650-5660), LY3214996 (International Publication No. WO 2016/106029 ), and MK-8353 (ACS Med Chem Lett 2018, 9(7): 761-767), and pharmacologically acceptable salts thereof.
  • In the present invention, the term "MEK inhibitor" refers to an agent that inhibits mitogen-activated extracellular signal-regulated kinase (MEK). The MEK inhibitor in the present invention may have an effect of inhibiting a kinase other than MEK. The MEK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include trametinib ( U.S. Patent No. 7378423 ), binimetinib ( U.S. Patent No. 7777050 ), selumetinib (International Publication No. WO 2005/023251 ), refametinib (International Publication No. WO 2007/014011 ), pimasertib (International Publication No. WO 2006/045514 ), cobimetinib ( U.S. Patent No. 7803839 ), E6201 (J Pharmacol Exp Ther 2009, 331(2): 485-95), PD-0325901 (Clin Cancer Res 2010, 16(8): 2450-7), RO5126766 (ACS Med Chem Lett 2014, 5(4): 309-14), and GDC-0623 (Bioorg Med Chem Lett 2014, 24(19): 4714-4723), and pharmacologically acceptable salts thereof.
  • Trametinib is preferably a dimethyl sulfoxide adduct (trametinib dimethyl sulfoxide).
  • In the present invention, the term "RAF inhibitor" refers to an agent that inhibits the kinase activity of RAF (preferably, BRAF). The RAF inhibitor in the present invention may have an effect of inhibiting a kinase other than RAF. The RAF inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib ( U.S. Patent No. 7351834 ), sorafenib ( U.S. Patent No. 7235576 ), vemurafenib ( U.S. Patent No. 7504509 ), dabrafenib ( U.S. Patent No. 7994185 ), encorafenib ( U.S. Patent No. 8501758 ), RAF265 (ACS Med Chem Lett 2015, 6(9): 961-5), GDC-5573 (International Publication No. WO 2013/100632 ), LY3009120 (J Med Chem 2015, 58(10): 4165-4179), and RO5126766, and pharmacologically acceptable salts thereof.
  • Regorafenib and pharmacologically acceptable salts thereof are each also known as a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, and an FGFR inhibitor. Sorafenib and pharmacologically acceptable salts thereof are each also known as a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, and an FLT3 inhibitor.
  • Regorafenib is preferably a hydrate (regorafenib hydrate). The pharmacologically acceptable salt of sorafenib is preferably a tosylate (sorafenib tosylate). The pharmacologically acceptable salt of dabrafenib is preferably a mesylate (dabrafenib mesylate).
  • In the present invention, the term "CDK1 inhibitor" refers to an agent that inhibits cyclin dependent kinase 1 (CDK1). The CDK1 inhibitor in the present invention may have an effect of inhibiting a kinase other than CDK1. The CDK1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include dinaciclib (International Publication No. WO 2005/077954 ), milciclib (J Med Chem 2009, 52(16): 5152-63), seliciclib (International Publication No. WO 1997/020842 ), alvocidib (International Publication No. WO 2001/053293 ), roniciclib (International Publication No. WO 2005/037800 ), voruciclib (International Publication No. WO 2007/148158 ), AT7519 (J Med Chem 2008, 51(16): 4986-4999), TP-1287, PHA-793887 (Bioorg Chem 2010, 18(5): 1844-53), KRX-0601, and rigosertib, and pharmacologically acceptable salts thereof.
  • Dinaciclib, seliciclib, alvocidib, voruciclib, AT7519, and PHA-793887 and pharmacologically acceptable salts thereof are each also known as a CDK2 inhibitor. Milciclib and pharmacologically acceptable salts thereof are each also known as a CDK2 inhibitor and a WEE1 inhibitor. Roniciclib and pharmacologically acceptable salts thereof are each also known as a CDK2 inhibitor and an Aurora kinase inhibitor.
  • In the present invention, the term "CDK2 inhibitor" refers to an agent that inhibits cyclin dependent kinase 2 (CDK2). The CDK2 inhibitor in the present invention may have an effect of inhibiting a kinase other than CDK2. The CDK2 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include CYC-065, PF-06873600, FN-1501, KRX-0601, dinaciclib, seliciclib, alvocidib, voruciclib, AT7519, PHA-793887, and roniciclib, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "CHK1 inhibitor" refers to an agent that inhibits checkpoint kinase 1 (CHK1). The CHK1 inhibitor in the present invention may have an effect of inhibiting a kinase other than CHK1. The CHK1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include MK-8776 (Bioorg Med Chem Lett 2011, 21(1): 471-4), LY2606368 (Mol Cancer Ther 2015, 14(9): 2004-13), LY2603618 (Invest New Drugs 2013, 31(1): 136-44), CBP501 (Mol Cancer Ther 2007, 6(1): 147-53), GDC-0425 (Org Process Res Dev 2015, 19(6): 661-672), CCT245737 (Oncotarget 2016, 7(3): 2329-42), and KRX-0601, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "WEE1 inhibitor" refers to an agent that inhibits Wee1-like protein kinase. The WEE1 inhibitor in the present invention may have an effect of inhibiting a kinase other than WEE1. The WEE1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include milciclib and MK-1775 (Mol Cancer Ther 2009, 8(11): 2992-3000), and pharmacologically acceptable salts thereof.
  • In the present invention, the term "PLK1 inhibitor" refers to an agent that inhibits Polo-like kinase 1 (PLK1). The PLK1 inhibitor in the present invention may have an effect of inhibiting a kinase other than PLK1. The PLK1 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include rigosertib and volasertib (International Publication No. WO 2012/049153 ), and pharmacologically acceptable salts thereof.
  • In the present invention, the term "Aurora kinase inhibitor" refers to an agent that inhibits Aurora kinase. The Aurora kinase inhibitor in the present invention may have an effect of inhibiting a kinase other than Aurora kinase. The Aurora kinase inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include alisertib (International Publication No. WO 2005/111039 ), ilorasertib (International Publication No. WO 2010/065825 ), ENMD-2076 (Clin Cancer Res 2011, 17(4): 849-60), AMG900 (Cancer Res 2010, 70(23): 9846-54), and roniciclib, and pharmacologically acceptable salts thereof.
  • Ilorasertib and pharmacologically acceptable salts thereof are each also known as a VEGFR inhibitor, a KIT inhibitor, a PDGFR inhibitor, an FLT3 inhibitor, and a CSF-1R inhibitor. ENMD-2076 is also known as a VEGFR inhibitor and an FLT3 inhibitor.
  • In the present invention, the term "Bcr-Abl inhibitor" refers to an agent that inhibits the tyrosine kinase Bcr-Abl. The Bcr-Abl inhibitor in the present invention may have an effect of inhibiting a kinase other than Bcr-Abl. The Bcr-Abl inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include imatinib ( U.S. Patent No. 6894051 ), dasatinib ( U.S. Patent No. 6596746 ), bosutinib ( U.S. Patent No. 6002008 ), nilotinib ( U.S. Patent No. 7169791 ), and ponatinib ( U.S. Patent No. 8114874 ), and pharmacologically acceptable salts thereof.
  • Imatinib and nilotinib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor and a PDGFR inhibitor. Dasatinib and pharmacologically acceptable salts thereof are each also known as an Src inhibitor, a KIT inhibitor, and a PDGFR inhibitor. Bosutinib and pharmacologically acceptable salts thereof are each also known as an Src inhibitor. Ponatinib and pharmacologically acceptable salts thereof are each also known as an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, and an FLT3 inhibitor.
  • The pharmacologically acceptable salt of imatinib is preferably mesylate (imatinib mesylate). Dasatinib is preferably a hydrate (dasatinib hydrate). Bosutinib is preferably a hydrate (bosutinib hydrate). The pharmacologically acceptable salt of nilotinib is preferably a hydrochloride hydrate (nilotinib hydrochloride hydrate). The pharmacologically acceptable salt of ponatinib is preferably a hydrochloride (ponatinib hydrochloride).
  • In the present invention, the term "Src inhibitor" refers to an agent that inhibits the protooncogene tyrosine protein kinase Src. The Src inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include dasatinib and bosutinib, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "EPH inhibitor" refers to an agent that inhibits tyrosine kinase of an erythropoietin-producing hepatocellular receptor (EPH). The EPH inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include dasatinib and ponatinib, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "VEGFR inhibitor" refers to an agent that inhibits tyrosine kinase of a vascular endothelial growth factor receptor (VEGFR). The VEGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than VEGFR tyrosine kinase. The VEGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, vandetanib ( U.S. Patent No. 7173038 ), sunitinib ( U.S. Patent No. 6573293 ), axitinib ( U.S. Patent No. 6534524 ), pazopanib ( U.S. Patent No. 7105530 ), lenvatinib ( U.S. Patent No. 7253286 ), nintedanib (hereinafter, also referred to as "BIBF1120") ( U.S. Patent No. 6762180 ), cabozantinib ( U.S. Patent No. 7579473 ), tivozanib (International Publication No. WO 2002/088110 ), brivanib (International Publication No. WO 2004/009784 ), linifanib (International Publication No. WO 2014/022975 ), lucitanib (International Publication No. WO 2008/112408 ), ilorasertib, and ENMD-2076, and pharmacologically acceptable salts thereof.
  • Vandetanib and pharmacologically acceptable salts thereof are each also known as an RET inhibitor. Sunitinib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FLT3 inhibitor, and a CSF-1R inhibitor. Axitinib and pharmacologically acceptable salts thereof are each also known as a PDGFR inhibitor, an FGFR inhibitor, and a CSF-1R inhibitor. Pazopanib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor, a PDGFR inhibitor, and an FGFR inhibitor. Lenvatinib and pharmacologically acceptable salts thereof are each also known as a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, and an FGFR inhibitor. Nintedanib and pharmacologically acceptable salts thereof are each also known as a PDGFR inhibitor and an FGFR inhibitor.
  • The pharmacologically acceptable salt of sunitinib is preferably a malate (sunitinib malate). The pharmacologically acceptable salt of pazopanib is preferably a hydrochloride (pazopanib hydrochloride). The pharmacologically acceptable salt of lenvatinib is preferably a mesylate (lenvatinib mesylate). The pharmacologically acceptable salt of nintedanib is preferably an ethanesulfonate (nintedanib ethanesulfonate). The pharmacologically acceptable salt of cabozantinib is preferably a malate (cabozantinib malate). The pharmacologically acceptable salt of tivozanib is preferably a hydrochloride hydrate (tivozanib hydrochloride hydrate).
  • In the present invention, the term "KIT inhibitor" refers to an agent that inhibits tyrosine kinase of the cytokine receptor KIT (c-kit, or also called CD117). The KIT inhibitor in the present invention may have an effect of inhibiting a kinase other than KIT tyrosine kinase. The KIT inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, and dasatinib, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "RET inhibitor" refers to an agent that inhibits RET (Rearranged during transfection) tyrosine kinase. The RET inhibitor in the present invention may have an effect of inhibiting a kinase other than RET tyrosine kinase. The RET inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, vandetanib, lenvatinib, and sunitinib, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "PDGFR inhibitor" refers to an agent that inhibits tyrosine kinase of a platelet-derived growth factor receptor (PDGFR). The PDGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than PDGFR tyrosine kinase. The PDGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, and dasatinib, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "FGFR inhibitor" refers to an agent that inhibits tyrosine kinase of a fibroblast growth factor receptor (FGFR). The FGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than FGFR tyrosine kinase. The FGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, and pazopanib, and pharmacologically acceptable salts thereof.
  • In the present invention, the term "BTK inhibitor" refers to an agent that inhibits Bruton's tyrosine kinase (BTK). The BTK inhibitor in the present invention may have an effect of inhibiting a kinase other than BTK. The BTK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ibrutinib ( U.S. Patent No. 7514444 ), acalabrutinib ( U.S. Patent No. 9290504 ), and tirabrutinib (International Publication No. WO 2011/152351 ), and pharmacologically acceptable salts thereof.
  • In the present invention, the term "FLT3 inhibitor" refers to an agent that inhibits FMS-like tyrosine kinase 3 (FLT3). The FLT3 inhibitor in the present invention may have an effect of inhibiting a kinase other than FLT3. The FLT3 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include gilteritinib ( U.S. Patent No. 7514444 ), quizartinib (J Med Chem 2009, 52(23): 7808-7816), midostaurin (International Publication No. WO 2003/037347 ), sorafenib, ilorasertib, ENMD-2076, and sunitinib, and pharmacologically acceptable salts thereof.
  • The pharmacologically acceptable salt of gilteritinib is preferably a fumarate (gilteritinib fumarate).
  • In the present invention, the term "ALK inhibitor" refers to an agent that inhibits anaplastic lymphoma kinase (ALK). The ALK inhibitor in the present invention may have an effect of inhibiting a kinase other than ALK. The ALK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include brigatinib ( U.S. Patent No. 9012462 ), crizotinib ( U.S. Patent No. 7858643 ), ceritinib ( U.S. Patent No. 7153964 ), alectinib ( U.S. Patent No. 9126931 ), and lorlatinib ( U.S. Patent No. 8680111 ), and pharmacologically acceptable salts thereof.
  • The pharmacologically acceptable salt of alectinib is preferably a hydrochloride (alectinib hydrochloride).
  • In the present invention, the term "JAK inhibitor" refers to an agent that inhibits Janus kinase (JAK). The JAK inhibitor in the present invention may have an effect of inhibiting a kinase other than JAK. The JAK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include ruxolitinib ( U.S. Patent No. 7598257 ), tofacitinib ( U.S. Patent No. 7265221 ), baricitinib ( U.S. Patent No. 8158616 ), and pacritinib (International Publication No. WO 2007/058627 ), and pharmacologically acceptable salts thereof.
  • The pharmacologically acceptable salt of ruxolitinib is preferably a phosphate (ruxolitinib phosphate). The pharmacologically acceptable salt of tofacitinib is preferably a citrate (tofacitinib citrate).
  • In the present invention, the term "MET inhibitor" refers to an agent that inhibits MET (also referred to as "c-Met"), a receptor-type tyrosine kinase whose ligand is a hepatocyte growth factor (HGF). The MET inhibitor in the present invention may have an effect of inhibiting a kinase other than MET. The MET inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include capmatinib (International Publication No. WO 2008/064157 ) and tepotinib (International Publication No. WO 2009/006959 ), and pharmacologically acceptable salts thereof.
  • In the present invention, the term "CSF-1R inhibitor" refers to an agent that inhibits tyrosine kinase of a colony-stimulating factor-1 receptor (CSF-1R). The CSF-1R inhibitor in the present invention may have an effect of inhibiting a kinase other than tyrosine kinase of CSF-1R. The CSF-1R inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include pexidartinib (Clin Cancer Res 2014, 20(12): 3146-58), BLZ-945 (International Publication No. WO 2007/121484 ), JNJ-40346527 (International Publication No. WO 2009/052237 ), JNJ-28312141 (J Med Chem 2011, 54(22): 7860-7883), ilorasertib, imatinib, sunitinib, and axitinib, and pharmacologically acceptable salts thereof.
  • The pharmacologically acceptable salt of pexidartinib is preferably a hydrochloride (pexidartinib hydrochloride).
  • In the present invention, the term "NTRK inhibitor" refers to an agent that inhibits neurotrophic tropomyosin receptor kinase (TRK). The NTRK inhibitor in the present invention may have an effect of inhibiting a kinase other than TRK. The NTRK inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include entrectinib (International Publication No. WO 2009/013126 ), and GR-389988 (International Publication No. WO 2015/089139 ), and pharmacologically acceptable salts thereof.
  • In the present invention, the term "EGFR inhibitor" refers to an agent that inhibits tyrosine kinase of an epidermal growth factor receptor (EGFR). The EGFR inhibitor in the present invention may have an effect of inhibiting a kinase other than tyrosine kinase of EGFR. The EGFR inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include gefitinib (International Publication No. WO 1996/033980 ), erlotinib (International Publication No. WO 1996/030347 ), afatinib (International Publication No. WO 2002/050043 ), osimertinib (International Publication No. WO 2013/014448 ), dacomitinib ( U.S. Patent No. 7772243 ), lapatinib ( U.S. Patent No. 713485 ), neratinib (J Med Chem 2005, 48(4): 1107-1131), pyrotinib (International Publication No. WO 2011/029265 ), and poziotinib (International Publication No. WO 2014/116070 ), and pharmacologically acceptable salts thereof.
  • In the present invention, the term "HER2 inhibitor" refers to an agent that inhibits tyrosine kinase of HER2 (human epidermal growth factor receptor type 2; ErbB-2). The HER2 inhibitor in the present invention may have an effect of inhibiting a kinase other than tyrosine kinase of HER2. The HER2 inhibitor in the present invention is not particularly limited as long as it is an agent that has the described characteristics, and preferred examples thereof can include tucatinib (International Publication No. WO 2013/056183 ), neratinib, mubritinib (International Publication No. WO 2001/077107 ), lapatinib, pyrotinib, and poziotinib, and pharmacologically acceptable salts thereof.
  • 5. Medicament
  • Described in the following are a pharmaceutical composition and a method of treatment wherein the antibody-drug conjugate according to the present invention and a kinase inhibitor are administered in combination.
  • The pharmaceutical composition and method of treatment of the present invention may be characterized in that the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times, or characterized in that the antibody-drug conjugate and the kinase inhibitor are contained as active components in a single formulation and administered.
  • In the pharmaceutical composition and method of treatment of the present invention, two or more of the kinase inhibitors used in the present invention can be administered in combination.
  • The pharmaceutical composition and method of treatment of the present invention can be used for treating cancer, and can be preferably used for treating at least one cancer selected from the group consisting of breast cancer (including triple-negative breast cancer and luminal breast cancer), gastric cancer (also called gastric adenocarcinoma), colorectal cancer (also called colon and rectal cancer, and including colon cancer and rectal cancer), lung cancer (including small cell lung cancer and non-small cell lung cancer), esophageal cancer, head-and-neck cancer (including salivary gland cancer and pharyngeal cancer), gastroesophageal junction adenocarcinoma, biliary tract cancer (including bile duct cancer), Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma, and can be more preferably used for treating at least one cancer selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, pancreatic cancer, kidney cancer, and ovarian cancer.
  • Among the antibody-drug conjugates used in the present invention, the kind of antibody preferably used in the antibody-drug conjugate can be determined by examining the type of cancer and tumor markers. For example, if HER2 expression is found in the cancer, an anti-HER2 antibody-drug conjugate can be preferably used; if HER3 expression is found in the cancer, an anti-HER3 antibody-drug conjugate can be preferably used; if TROP2 expression is found in the cancer, an anti-TROP2 antibody-drug conjugate can be preferably used; if B7-H3 expression is found in the cancer, an anti-B7-H3 antibody-drug conjugate can be preferably used; and if CDH6 expression is found in the cancer, an anti-CDH6 antibody-drug conjugate can be preferably used.
  • The presence or absence of HER2, HER3, TROP2, B7-H3, and CDH6, and other tumor markers can be checked by, for example, collecting tumor tissue from a cancer patient, and subjecting the formalin-fixed paraffin-embedded specimen (FFPE) to an examination at a gene product (protein) level, such as an immunohistochemistry (IHC) method, a flow cytometry, a western blot method, or an examination at a gene transcription level such as an in situ hybridization method (ISH), a quantitative PCR method (q-PCR), or a microarray analysis; alternatively, it can also be checked by collecting cell-free blood circulating tumor DNA (ctDNA) from a cancer patient and subjecting it to an examination which uses a method such as next generation sequencing (NGS).
  • The pharmaceutical composition and method of treatment of the present invention can be preferably used for a mammal, and can be more preferably used for a human.
  • The antitumor effect of the pharmaceutical composition and method of treatment of the present invention can be confirmed by, for example, generating a model in which cancer cells are transplanted to a test animal, and measuring reduction in tumor volume or lifeprolonging effects due to applying the pharmaceutical composition and method of treatment of the present invention. Furthermore, comparison with the antitumor effect of single administrations of each of the antibody-drug conjugate and the kinase inhibitor used in the present invention can provide confirmation of the combined effect of the antibody-drug conjugate and the kinase inhibitor used in the present invention.
  • In addition, the antitumor effect of the pharmaceutical composition and method of treatment of the present invention can be confirmed, in a clinical study, with the Response Evaluation Criteria in Solid Tumors (RECIST) evaluation method, WHO's evaluation method, Macdonald's evaluation method, measurement of body weight, and other methods; and can be determined by indicators such as Complete response (CR), Partial response (PR), Progressive disease (PD), Objective response rate (ORR), Duration of response (DoR), Progression-free survival (PFS), and Overall survival (OS).
  • The foregoing methods can provide confirmation of superiority in terms of the antitumor effect of the pharmaceutical composition and method of treatment of the present invention compared to existing pharmaceutical compositions and methods of treatment for cancer therapy.
  • The pharmaceutical composition and method of treatment of the present invention can retard growth of cancer cells, suppress their proliferation, and further can kill cancer cells. These effects can allow cancer patients to be free from symptoms caused by cancer or can achieve an improvement in the QOL of cancer patients and attain a therapeutic effect by sustaining the lives of the cancer patients. Even if the pharmaceutical composition and method of treatment of the present invention do not accomplish the killing of cancer cells, they can achieve higher QOL of cancer patients while achieving longer-term survival, by inhibiting or controlling the growth of cancer cells.
  • The pharmaceutical composition of the present invention can be expected to exert a therapeutic effect by application as systemic therapy to patients, and additionally, by local application to cancer tissues.
  • The pharmaceutical composition of the present invention may be administered as a pharmaceutical composition containing at least one pharmaceutically suitable ingredient. The pharmaceutically suitable ingredient can be suitably selected and applied from formulation additives or the like that are generally used in the art, in view of the dosage, administration concentration, or the like of the antibody-drug conjugate and the kinase inhibitor used in the present invention. For example, the antibody-drug conjugate used in the present invention may be administered as a pharmaceutical composition containing a buffer such as histidine buffer, an excipient such as sucrose and trehalose, and a surfactant such as Polysorbates 80 and 20. The pharmaceutical composition containing the antibody-drug conjugate used in the present invention can be preferably used as an injection, can be more preferably used as an aqueous injection or a lyophilized injection, and can be even more preferably used as a lyophilized injection.
  • In the case that the pharmaceutical composition containing the antibody-drug conjugate used in the present invention is an aqueous injection, the aqueous injection can be preferably diluted with a suitable diluent and then given as an intravenous infusion. For the diluent, a dextrose solution, physiological saline, and the like, can be exemplified, and a dextrose solution can be preferably exemplified, and a 5% dextrose solution can be more preferably exemplified.
  • In the case that the pharmaceutical composition containing the antibody-drug conjugate used in the present invention is a lyophilized injection, it can be preferably dissolved in water for injection, subsequently a required amount can be diluted with a suitable diluent and then given as an intravenous infusion. For the diluent, a dextrose solution, physiological saline, and the like, can be exemplified, and a dextrose solution can be preferably exemplified, and a 5% dextrose solution can be more preferably exemplified.
  • Examples of the administration route which may be used to administer the pharmaceutical composition of present invention include intravenous, intradermal, subcutaneous, intramuscular, and intraperitoneal routes; and preferably include an intravenous route.
  • The antibody-drug conjugate used in the present invention can be administered to a human once at intervals of 1 to 180 days, and can be preferably administered once a week, once every 2 weeks, once every 3 weeks, or once every 4 weeks, and can be even more preferably administered once every 3 weeks. Also, the antibody-drug conjugate used in the present invention can be administered at a dose of about 0.001 to 100 mg/kg, and can be preferably administered at a dose of 0.8 to 12.4 mg/kg. In the case that the antibody-drug conjugate used in the present invention is an anti-HER2 antibody-drug conjugate, it can be preferably administered once every 3 weeks at a dose of 0.8 mg/kg, 1.6 mg/kg, 3.2 mg/kg, 5.4 mg/kg, 6.4 mg/kg, 7.4 mg/kg, or 8 mg/kg. In the case that the antibody-drug conjugate used in the present invention is an anti-HER3 antibody-drug conjugate, it can be preferably administered once every 3 weeks at a dose of 1.6 mg/kg, 3.2 mg/kg, 4.8 mg/kg, 6.4 mg/kg, 8.0 mg/kg, 9.6 mg/kg, or 12.8 mg/kg. In the case that the antibody-drug conjugate used in the present invention is an anti-TROP2 antibody-drug conjugate, it can be preferably administered once every 3 weeks at a dose of 0.27 mg/kg, 0.5 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 4.0 mg/kg, 6.0 mg/kg, or 8.0 mg/kg.
  • The kinase inhibitor according to the present invention can be orally administered to a human once or twice at intervals of 1 to 7 days, and can be preferably orally administered once a day or twice per day. Also, the kinase inhibitor used in the present invention can be orally administered at a dose of 0.1 mg to 3000 mg, and can be preferably administered at a dose of 2.5 mg to 600 mg. Furthermore, the kinase inhibitor used in the present invention can be administered to a human as an intravenous drip once at intervals of 1 to 180 days, and can be preferably administered as an intravenous drip once a week, once every 2 weeks, once every 3 weeks, or once every 4 weeks. Also, the kinase inhibitor used in the present invention can be administered as an intravenous drip at a dose of 0.1 mg to 3000 mg, and can be preferably administered as an intravenous drip at a dose of 10 mg to 100 mg.
  • In the case that the kinase inhibitor used in the present invention is abemaciclib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 50 mg, 100 mg, 150 mg, or 200 mg.
  • In the case that the kinase inhibitor used in the present invention is palbociclib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 75 mg, 100 mg, or 125 mg.
  • In the case that the kinase inhibitor used in the present invention is ribociclib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 200 mg, 400 mg, or 600 mg.
  • In the case that the kinase inhibitor used in the present invention is everolimus or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 2.5 mg, 5 mg, 7.5 mg, 10 mg, 15 mg, or 20 mg.
  • In the case that the kinase inhibitor used in the present invention is sirolimus or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 0.5 mg, 1 mg, 1.5 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 8 mg, 10 mg, 12 mg, or 15 mg.
  • In the case that the kinase inhibitor used in the present invention is temsirolimus or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably administered as an intravenous drip once a week at a dose of 25 mg.
  • In the case that the kinase inhibitor used in the present invention is copanlisib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably administered as an intravenous drip once with intervals of 1 to 3 weeks at a dose of 60 mg.
  • In the case that the kinase inhibitor used in the present invention is idelalisib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 100 mg or 150 mg.
  • In the case that the kinase inhibitor used in the present invention is duvelisib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 15 mg or 25 mg.
  • In the case that the kinase inhibitor used in the present invention is trametinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 0.5 mg, 1 mg, 1.5 mg, or 2 mg.
  • In the case that the kinase inhibitor used in the present invention is binimetinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 15 mg, 30 mg, or 45 mg.
  • In the case that the kinase inhibitor used in the present invention is cobimetinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 40 mg, or 60 mg.
  • In the case that the kinase inhibitor used in the present invention is regorafenib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg, 80 mg, 120 mg, or 160 mg.
  • In the case that the kinase inhibitor used in the present invention is sorafenib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 200 mg or 400 mg.
  • In the case that the kinase inhibitor used in the present invention is vemurafenib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 240 mg, 480 mg, 720 mg, or 960 mg.
  • In the case that the kinase inhibitor used in the present invention is dabrafenib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 50 mg, 75 mg, 100 mg, 125 mg, or 150 mg.
  • In the case that the kinase inhibitor used in the present invention is encorafenib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 50 mg, 75 mg, 100 mg, 125 mg, 150 mg, 175 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, or 450 mg.
  • In the case that the kinase inhibitor used in the present invention is imatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg, or twice per day at a dose of 400 mg.
  • In the case that the kinase inhibitor used in the present invention is dasatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 50 mg, 70 mg, 80 mg, 100 mg, or 140 mg.
  • In the case that the kinase inhibitor used in the present invention is bosutinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg.
  • In the case that the kinase inhibitor used in the present invention is nilotinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 150 mg, 200 mg, 300 mg, or 400 mg.
  • In the case that the kinase inhibitor used in the present invention is ponatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 15 mg, 30 mg, or 45 mg.
  • In the case that the kinase inhibitor used in the present invention is vandetanib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 100 mg, 200 mg, or 300 mg.
  • In the case that the kinase inhibitor used in the present invention is axitinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 1 mg, 2 mg, 3 mg, 4 mg, or 5 mg.
  • In the case that the kinase inhibitor used in the present invention is pazopanib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 200 mg, 400 mg, 600 mg, or 800 mg.
  • In the case that the kinase inhibitor used in the present invention is lenvatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 4 mg, 8 mg, 10 mg, 12 mg, 14 mg, 16 mg, 18 mg, 20 mg, or 24 mg.
  • In the case that the kinase inhibitor used in the present invention is nintedanib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 100 mg or 150 mg.
  • In the case that the kinase inhibitor used in the present invention is cabozantinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 40 mg, or 60 mg.
  • In the case that the kinase inhibitor used in the present invention is ibrutinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 70 mg, 140 mg, 210 mg, 280 mg, 350 mg, 420 mg, 490 mg, or 560 mg.
  • In the case that the kinase inhibitor used in the present invention is acalabrutinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once per 12 hours at a dose of 100 mg.
  • In the case that the kinase inhibitor used in the present invention is gilteritinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg, 80 mg, or 120 mg.
  • In the case that the kinase inhibitor used in the present invention is midostaurin or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 25 mg, 50 mg, 75 mg, or 100 mg.
  • In the case that the kinase inhibitor used in the present invention is brigatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 30 mg, 60 mg, 90 mg, 120 mg, 150 mg, or 180 mg.
  • In the case that the kinase inhibitor used in the present invention is crizotinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 200 mg or 250 mg.
  • In the case that the kinase inhibitor used in the present invention is ceritinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 150 mg, 300 mg, 450 mg, 600 mg, or 750 mg.
  • In the case that the kinase inhibitor used in the present invention is alectinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 150 mg, 300 mg, 450 mg, or 600 mg.
  • In the case that the kinase inhibitor used in the present invention is lorlatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 25 mg, 50 mg, 75 mg, or 100 mg.
  • In the case that the kinase inhibitor used in the present invention is ruxolitinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 5 mg, 10 mg, 15 mg, 20 mg, or 25 mg.
  • In the case that the kinase inhibitor used in the present invention is tofacitinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered twice per day at a dose of 5 mg or 10 mg per administration, and in using as a sustained-release formulation preferably administered once a day at a dose of 11 mg.
  • In the case that the kinase inhibitor used in the present invention is baricitinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 2 mg.
  • In the case that the kinase inhibitor used in the present invention is gefitinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 250 mg.
  • In the case that the kinase inhibitor used in the present invention is erlotinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 25 mg, 50 mg, 75 mg, 100 mg, 125 mg, or 150 mg.
  • In the case that the kinase inhibitor used in the present invention is afatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 20 mg, 30 mg, or 40 mg.
  • In the case that the kinase inhibitor used in the present invention is osimertinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg or 80 mg.
  • In the case that the kinase inhibitor used in the present invention is dacomitinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 15 mg, 30 mg, or 45 mg.
  • In the case that the kinase inhibitor used in the present invention is lapatinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 250 mg, 500 mg, 750 mg, 1000 mg, 1250 mg, or 1500 mg.
  • In the case that the kinase inhibitor used in the present invention is neratinib or a pharmacologically acceptable salt thereof, the kinase inhibitor can be preferably orally administered once a day at a dose of 40 mg, 80 mg, 120 mg, 160 mg, 200 mg, or 240 mg.
  • The pharmaceutical composition and method of treatment of the present invention may further contain a cancer therapeutic agent other than the antibody-drug conjugate and the kinase inhibitor according to the present invention. The pharmaceutical composition and method of treatment of the present invention can also be administered in combination with another cancer therapeutic agent, thereby enhancing the antitumor effect. Other cancer therapeutic agents to be used for such purpose may be administered to a subject simultaneously with, separately from, or subsequently with the pharmaceutical composition of the present invention, or may be administered while varying the dosage interval for each. Such cancer therapeutic agents are not limited as long as they are agents having antitumor activity, and can be exemplified by at least one selected from the group consisting of irinotecan (CPT-11), cisplatin, carboplatin, oxaliplatin, fluorouracil (5-FU), gemcitabine, capecitabine, paclitaxel, docetaxel, doxorubicin, epirubicin, cyclophosphamide, mitomycin C, a tegafur-gimeracil-oteracil combination drug, cetuximab, panitumumab, bevacizumab, ramucirumab, a trifluridinetipiracil combination drug, methotrexate, pemetrexed, tamoxifen, toremifene, fulvestrant, leuprorelin, goserelin, letrozole, anastrozole, a progesterone formulation, trastuzumab, and pertuzumab.
  • The pharmaceutical composition and method of treatment of the present invention can also be used in combination with radiotherapy. For example, a cancer patient may receive radiotherapy before and/or after or simultaneously with receiving therapy with the pharmaceutical composition of the present invention.
  • The pharmaceutical composition and method of treatment of the present invention can also be used as adjuvant chemotherapy in combination with a surgical procedure. The pharmaceutical composition of the present invention may be administered for the purpose of diminishing the size of a tumor before surgical procedure (referred to as pre-operative adjuvant chemotherapy or neoadjuvant therapy), or may be administered after a surgical procedure for the purpose of preventing the recurrence of a tumor (referred to as post-operative adjuvant chemotherapy or adjuvant therapy).
  • Examples
  • The present invention is specifically described in view of the examples shown below. However, the present invention is not limited to these. Further, it is by no means to be interpreted in a limited way.
  • Example 1: Production of antibody-drug conjugate (1)
  • In accordance with a production method described in International Publication No. WO 2015/115091 with use of an anti-HER2 antibody (an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2), an antibody-drug conjugate in which a drug-linker represented by the following formula:
    Figure imgb0036
    Figure imgb0037
    wherein A represents a connecting position to an antibody, is conjugated to the anti-HER2 antibody via a thioether bond (hereinafter, referred to as the "antibody-drug conjugate (1)") was produced. The DAR of the antibody-drug conjugate (1) is 7.7 or 7.8.
  • Example 2: Antitumor test (1)
  • Mouse: 5 to 6-week-old BALB/c nude mice (Charles River Laboratories Japan, Inc.) were subjected to experiment.
  • Assay and calculation expression: In all of the studies, the major axis and minor axis of a tumor were measured twice a week by using an electronic digital caliper (CD15-CX, Mitutoyo Corp.), and the tumor volume (mm3) was calculated. The calculation expression is as shown below. Tumor volume mm 3 = 1 / 2 × Major axis mm × Minor axis mm 2
    Figure imgb0038
  • The antibody-drug conjugate (1) was diluted with ABS buffer (10 mM acetate buffer solution (pH 5.5), 5% sorbitol), and intravenously administered to the tail vein at a liquid volume of 10 mL/kg. Everolimus was suspended in 30% propylene glycol and 5% Tween 80, and orally administered at a liquid volume of 10 mL/kg. Taselisib was suspended in 0.5% methyl cellulose and 0.2% Tween 80, and orally administered at a liquid volume of 10 mL/kg. Abemaciclib was suspended in 1% hydroxyethyl cellulose and 0.1% antifoam/25 mM phosphate buffer, pH2, and orally administered at a liquid volume of 10 mL/kg.
  • MDA-MB-453 cells, a human breast cancer cell line, purchased from ATCC (American Type Culture Collection) were suspended in Matrigel basement membrane matrix, and 1 × 107 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0). The antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 0.5 mg/kg on Day 0. Everolimus was administered once a day, five times a week, at a dose of 5 mg/kg for 3 weeks. Taselisib was administered once a day, five times a week, at a dose of 5 mg/kg for 3 weeks. Abemaciclib was administered once a day, five times a week, at a dose of 30 mg/kg for 2 weeks. Single administration groups and a combined administration group with the antibody-drug conjugate (1) and each kinase inhibitor, and a solvent administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (1) and everolimus are shown in Figure 11. The tumor growth inhibition (TGI) of single administration of everolimus on the day of effect determination was 64%. The TGI of single administration of the antibody-drug conjugate (1) was 75%. For combined administration of the antibody-drug conjugate (1) and everolimus, on the other hand, tumor growth inhibitory effect significantly superior to that of single administration of everolimus was found (P = 0.0059 (calculated with Dunnett's test, the same applies hereinafter)). The tumor growth inhibition (TGI: 89%) was higher than those of single administration of them, demonstrating strong effect of their combined use. In the figure, the abscissa depicts days from the initiation of administration, and the ordinate depicts tumor volumes. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group. In evaluation examples below for antitumor tests, the tests were conducted with the procedure used in this evaluation example, unless otherwise stated.
  • Results of combined use of the antibody-drug conjugate (1) and taselisib are shown in Figure 12. The TGI of single administration of taselisib was 67%. The TGI of single administration of the antibody-drug conjugate (1) was 75%. For combined administration of the antibody-drug conjugate (1) and taselisib, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (1) and single administration of taselisib was found (P = 0.0039 and P = 0.0043, respectively). The tumor growth inhibition (TGI: 92%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Results of combined use of the antibody-drug conjugate (1) and abemaciclib are shown in Figure 13. The TGI of single administration of abemaciclib was 68%. The TGI of single administration of the antibody-drug conjugate (1) was 75%. For combined administration of the antibody-drug conjugate (1) and abemaciclib, on the other hand, tumor growth inhibitory effect significantly superior to that of single administration of abemaciclib was found (P = 0.0429). The tumor growth inhibition (TGI: 87%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 3: Antitumor test (2)
  • JIMT-1 cells, a human breast cancer cell line, purchased from DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH) were suspended in physiological saline, and 5 × 106 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 15 days after the transplantation (Day 0). The antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 10 mg/kg on Day 0. Everolimus was administered once a day, five times a week and seven times in total, at a dose of 5 mg/kg. Single administration groups and a combined administration group with the antibody-drug conjugate (1) and everolimus, and a solvent administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (1) and everolimus are shown in Figure 14. The TGI of single administration of everolimus was 52%. The TGI of single administration of the antibody-drug conjugate (1) was 53%. For combined administration of the antibody-drug conjugate (1) and everolimus, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (1) and single administration of everolimus was found (P = 0.0131 and P = 0.0054, respectively). The tumor growth inhibition (TGI: 93%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 4: Antitumor test (3)
  • COLO201 cells, a human colorectal cancer cell line, purchased from ATCC were suspended in 50% Matrigel solution, and 5 × 106 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0). The antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 3 mg/kg on Day 0. Regorafenib was dissolved in Cremophor EL/95% ethanol and then diluted with distilled water, and orally administered at a liquid volume of 10 mL/kg. Administration was performed once a day, five times a week, at a dose of 10 mg/kg for three weeks. Single administration groups and a combined administration group with the antibody-drug conjugate (1) and regorafenib, and a solvent administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (1) and regorafenib are shown in Figure 15. The TGI of single administration of regorafenib was 81%. The TGI of single administration of the antibody-drug conjugate (1) was 67%. For combined administration of the antibody-drug conjugate (1) and regorafenib, on the other hand, tumor growth inhibitory effect significantly superior to that of single administration of the antibody-drug conjugate (1) was found (P = 0.0476). The tumor growth inhibition (TGI: 99%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 5: Antitumor test (4)
  • KPL-4 cells, a human breast cancer cell line, obtained from Dr. Junichi Kurebayashi, Kawasaki Medical School (British Journal of Cancer. (1999)79 (5/6). 707-717) were suspended in physiological saline, and 1.5 × 107 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 17 days after the transplantation (Day 0). The antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 7.5 mg/kg on Day 0. Tucatinib was suspended in 0.5% methyl cellulose, and orally administered at a liquid volume of 10 mL/kg. Administration was performed once a day, five times a week, at a dose of 100 mg/kg for two weeks. Single administration groups and a combined administration group with the antibody-drug conjugate (1) and tucatinib, and a solvent administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (1) and tucatinib are shown in Figure 16. The TGI of single administration of tucatinib was 78%. The TGI of single administration of the antibody-drug conjugate (1) was 78%. For combined administration of the antibody-drug conjugate (1) and tucatinib, on the other hand, the tumor growth inhibition (TGI: 96%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 6: Antitumor test (5)
  • MDA-MB-453 cells, a human breast cancer cell line, purchased from ATCC (American Type Culture Collection) were suspended in Matrigel basement membrane matrix, and 1 × 107 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0). The antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 0.5 mg/kg on Day 0. Tucatinib was administered once a day, five times a week, at a dose of 100 mg/kg for three weeks. Single administration groups and a combined administration group with the antibody-drug conjugate (1) and tucatinib, and a solvent administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (1) and tucatinib are shown in Figure 17. The TGI of single administration of tucatinib was 36%. The TGI of single administration of the antibody-drug conjugate (1) was 52%. For combined administration of the antibody-drug conjugate (1) and tucatinib, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (1) and single administration of tucatinib was found (P = 0.019 and P = 0.001, respectively). The tumor growth inhibition (TGI: 85%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 7: Antitumor test (6)
  • NCI-N87 cells, a human gastric cancer cell line, purchased from ATCC (American Type Culture Collection) were suspended in physiological saline, and 1 × 107 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 6 days after the transplantation (Day 0). The antibody-drug conjugate (1) was intravenously administered to the tail vein of each mouse at a dose of 2 mg/kg on Day 0. Tucatinib was administered once a day, five times a week, at a dose of 100 mg/kg for two weeks. Single administration groups and a combined administration group with the antibody-drug conjugate (1) and tucatinib, and a solvent administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (1) and tucatinib are shown in Figure 18. The TGI of single administration of tucatinib was 65%. The TGI of single administration of the antibody-drug conjugate (1) was 71%. For combined administration of the antibody-drug conjugate (1) and tucatinib, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (1) and single administration of tucatinib was found (P = 0.0199 and P = 0.0034, respectively). The tumor growth inhibition (TGI: 92%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 8: Production of antibody-drug conjugate (2)
  • In accordance with a production method described in International Publication No. WO 2015/155998 with use of an anti-HER3 antibody (an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4), an antibody-drug conjugate in which a drug-linker represented by the following formula:
    Figure imgb0039
    • wherein A represents a connecting position to an anti-HER3 antibody,
    • is conjugated to the anti-HER3 antibody via a thioether bond (hereinafter, referred to as the "antibody-drug conjugate (2)") was produced. The DAR of the antibody-drug conjugate (2) is 7 to 8.
    Example 9: Antitumor test (7)
  • KPL-4 cells, a human breast cancer cell line, obtained from Dr. Junichi Kurebayashi, Kawasaki Medical School (British Journal of Cancer. (1999)79 (5/6). 707-717) were suspended in physiological saline, and 1.5 × 107 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 17 days after the transplantation (Day 0). The antibody-drug conjugate (2) was diluted with ABS buffer, and intravenously administered to the tail vein of each mouse once a week at a dose of 10 mg/kg (a liquid volume of 10 mL/kg) for three weeks ( Days 0, 7, 14). Neratinib was suspended in 0.5% methyl cellulose, and orally administered once a day, five times a week, at a dose of 20 mg/kg (a liquid volume of 10 mL/kg) for two weeks. Single administration groups and a combined administration group with the antibody-drug conjugate (2) and neratinib, and an ABS administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (2) and neratinib are shown in Figure 19. The TGI of single administration of neratinib on the day of effect determination was 84%. The TGI of single administration of the antibody-drug conjugate (2) was 67%. For combined administration of the antibody-drug conjugate (2) and neratinib, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (2) and single administration of neratinib was found (P < 0.0001 and P = 0.0098, respectively). The tumor growth inhibition (TGI: 95%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 10: Production of antibody-drug conjugate (3)
  • In accordance with a production method described in International Publication No. WO 2018/212136 with use of an anti-CDH6 antibody (an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10), an antibody-drug conjugate in which a drug-linker represented by the following formula:
    Figure imgb0040
    • wherein A represents a connecting position to an anti-CDH6 antibody,
    • is conjugated to the anti-CDH6 antibody via a thioether bond (hereinafter, referred to as the "antibody-drug conjugate (3)") was produced. The DAR of the antibody-drug conjugate (3) is 7 to 8.
    Example 11: Antitumor test (8)
  • Suspended in 100% Matrigel were 786-O cells, a human kidney cancer cell line, purchased from ATCC, and 4 × 106 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 19 days after the transplantation (Day 0). The antibody-drug conjugate (3) was intravenously administered to the tail vein of each mouse at a dose of 10 mg/kg on Day 0. Cabozantinib was administered once a day, five times a week, 20 times in total, at a dose of 40 mg/kg. Single administration groups and a combined administration group with the antibody-drug conjugate (3) and cabozantinib, and a solvent administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (3) and cabozantinib are shown in Figure 20. The TGI of single administration of cabozantinib was 67%. The TGI of single administration of the antibody-drug conjugate (3) was 71%. For combined administration of the antibody-drug conjugate (3) and cabozantinib, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (3) and single administration of cabozantinib was found (P = 0.0024 and P = 0.0004, respectively). The tumor growth inhibition (TGI: 90%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 12: Production of antibody-drug conjugate (4)
  • In accordance with a production method described in International Publication No. WO 2015/098099 and International Publication No. WO 2017/002776 with use of an anti-TROP2 antibody (an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6), an antibody-drug conjugate in which a drug-linker represented by the following formula:
    Figure imgb0041
    • wherein A represents a connecting position to an anti-TROP2 antibody,
    • is conjugated to the anti-TROP2 antibody via a thioether bond (hereinafter, referred to as the "antibody-drug conjugate (4)") was produced. The DAR of the antibody-drug conjugate (4) is 3.5 or 4.5.
    Example 13: Production of Compound (1)
  • In accordance with a production method described in International Publication No. WO 2014/057687 and International Publication No. WO 2015/115091 , a compound represented by the following formula:
    Figure imgb0042
    (hereinafter, referred to as the "Compound (1)") was produced.
  • Example 14: Cell growth inhibition test (1)
  • The human gastric cancer cell line NCI-N87 purchased from ATCC was used for evaluation. To a 1536-well cell culture plate, erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with dimethyl sulfoxide (DMSO) to concentrations of 2 mM, 400 µM, 80 µM, 16 µM, 3.2 µM, and 640 nM, or DMSO was added at 25 nL/well. Further, Compound (1) prepared with an RPMI1640 Medium (Thermo Fisher Scientific) containing 10% fetal bovine serum (GE Healthcare) to concentrations of 60 nM, 24 nM, 9.6 nM, 3.8 nM, 1.5 nM, and 0.61 nM, or the antibody-drug conjugate (4) prepared to concentrations of 32 nM, 11 nM, 3.6 nM, 1.2 nM, 0.40 nM, and 0.13 nM, or the antibody-drug conjugate (1) prepared to concentrations of 8.0 nM, 2.7 nM, 0.89 nM, 0.30 nM, 0.10 nM, and 0.033 nM was added at 2.5 µL/well. Subsequently, NCI-N87 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 4 × 104 cells/mL were seeded at 2.5 µL/well, and cultured at 37°C under 5%CO2 for 6 days.
  • After culturing, a solution obtained by diluting CellTiter-Glo 2.0 Assay (Promega Corporation) with an equivalent amount of Glo Lysis buffer, 1 × (Promega Corporation) was added at 2 µL/well, and incubation was performed at room temperature for 1 hour, and luminescence intensity was then measured for each well.
  • Cell growth inhibition rate (%) was calculated by using the following calculation expression. Cell growth inhibition rate % = 100 × T B / C B 100
    Figure imgb0043
    • T: Mean luminescence intensity of wells with specimen
    • B: Mean luminescence intensity of wells with DMSO and medium
    • C: Mean luminescence intensity of wells with DMSO and cells
  • Sigmoid fitting with Genedata Screener Analyzer Version 14 (Genedata AG, hereinafter referred to as Screener) was performed for concentration-dependent transition of cell growth inhibition rates under each of combined use conditions.
  • For effect of combined use, differences between estimated values of additive effect by a Loewe model (Greco WR. et al., Pharmacol. Rev. 1995 Jun; 47 (2): 331-85) and sigmoid-fitted cell growth inhibition rates (%) were matrixized, and a Synergy Score was calculated from the matrix components by using a procedure demonstrated in the literature (Lehar J. et al., Nat Biotechnol. 2009 Jul; 27 (7): 659-66). Synergy Score = 0 indicates additive action, Synergy Score > 0 indicates synergistic action, and Synergy Score < 0 indicates antagonistic action.
  • Synergy Scores for the combinations are shown in Table 1. In the cell growth inhibition test for the NCI-N87 cell line, Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride) or brigatinib, the antibody-drug conjugate (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120. [Table 1]
    Synergy Scores for each combination in NCI-N87 cell line
    Erlotinib Poziotinib Mubritinib Brigatinib BIBF1120
    Compound (1) 0.48 -0.53 -0.45 0.11 -0.44
    Antibody-drug conjugate (1) 2.38 1.10 -0.06 1.55 1.73
    Antibody-drug conjugate (4) 2.33 1.50 1.67 0.41 1.16
  • Example 15: Cell growth inhibition test (2)
  • The human breast cancer cell line KPL-4 obtained from Dr. Junichi Kurebayashi, Kawasaki Medical School (British Journal of Cancer. (1999)79 (5/6). 707-717) was used for evaluation. To a 1536-well cell culture plate, erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 µM, 80 µM, 16 µM, 3.2 µM, and 640 nM, or DMSO was added at 25 nL/well. Further, Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 36 nM, 20 nM, 11 nM, 6.2 nM, 3.4 nM, and 1.9 nM, or the antibody-drug conjugate (4) prepared to concentrations of 40 nM, 13 nM, 4.4 nM, 1.5 nM, 0.49 nM, and 0.16 nM, or the antibody-drug conjugate (1) prepared to concentrations of 3.7 nM, 1.3 nM, 0.48 nM, 0.17 nM, 0.061 nM, and 0.022 nM was added at 2.5 µL/well. Subsequently, KPL-4 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 1 × 104 cells/mL were seeded at 2.5 µL/well, and cultured at 37°C under 5%CO2 for 6 days. After culturing, a solution obtained by diluting CellTiter-Glo 2.0 Assay with an equivalent amount of Glo Lysis buffer, 1 × was added at 2 µL/well, and incubation was performed at room temperature for 1 hour, and luminescence intensity was then measured for each well.
  • Analysis of cell growth inhibition rates (%) and effect of combined use under each of the conditions was performed in the same manner as in Example 14.
  • Synergy Scores for the combinations are shown in Table 2. In the cell growth inhibition test for the KPL-4 cell line, Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), mubritinib, or BIBF1120, the antibody-drug conjugate (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), mubritinib, brigatinib, or BIBF1120. [Table 2]
    Synergy Scores for each combination in KPL-4 cell line
    Erlotinib Poziotinib Mubritinib Brigatinib BIBF1120
    Compound (1) 0.22 -0.38 0.57 -0.02 0.99
    Antibody-drug conjugate (1) 0.77 0.03 0.28 1.58 2.26
    Antibody-drug conjugate (4) 3.51 -0.29 1.35 1.15 1.24
  • Example 16: Cell growth inhibition test (3)
  • The human lung cancer cell line EBC-1 obtained from Health Science Research Resources Bank (currently known as Japanese Collection of Research Bioresources (JCRB) Cell Bank) was used for evaluation. To a 1536-well cell culture plate, erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 µM, 80 µM, 16 µM, 3.2 µM, and 640 nM or DMSO was added at 25 nL/well. Further, Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 16 nM, 8.0 nM, 4.0 nM, 2.0 nM, 1.0 nM, and 0.50 nM, or the antibody-drug conjugate (4) prepared to concentrations of 40 nM, 13 nM, 4.4 nM, 1.5 nM, 0.49 nM, and 0.16 nM was added at 2.5 µL/well. Subsequently, EBC-1 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 2 × 104 cells/mL were seeded at 2.5 µL/well, and cultured at 37°C under 5%CO2 for 6 days. After culturing, a solution obtained by diluting CellTiter-Glo 2.0 Assay with an equivalent amount of Glo Lysis buffer, 1 × was added at 2 µL/well, and incubation was performed at room temperature for 1 hour, and luminescence intensity was then measured for each well. Analysis of cell growth inhibition rates (%) and effect of combined use under each of the conditions was performed in the same manner as in Example 14.
  • Synergy Scores for the combinations are shown in Table 3.
  • In the cell growth inhibition test for the EBC-1 cell line, Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120. [Table 3]
    Synergy Scores for each combination in EBC-1 cell line
    Erlotinib Poziotinib Mubritinib Brigatinib BIBF1120
    Compound (1) 1.88 0.45 -0.60 0.35 0.50
    Antibody-drug conjugate (4) 6.96 1.42 0.72 2.52 6.04
  • Example 17: Cell growth inhibition test (4)
  • The human breast cancer cell line HCC70 obtained from ATCC was used for evaluation. To a 1536-well cell culture plate, erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 µM, 80 µM, 16 µM, 3.2 µM, and 640 nM or DMSO was added at 25 nL/well. Further, Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 800 nM, 200 nM, 50 nM, 13 nM, 3.1 nM, and 0.78 nM, or the antibody-drug conjugate (4) prepared to concentrations of 5.6 nM, 2.0 nM, 0.71 nM, 0.26 nM, 0.091 nM, and 0.033 nM was added at 2.5 µL/well. Subsequently, HCC70 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 4 × 104 cells/mL were seeded at 2.5 µL/well, and cultured at 37°C under 5%CO2 for 6 days. After culturing, a solution obtained by diluting CellTiter-Glo 2.0 Assay with an equivalent amount of Glo Lysis buffer, 1 × was added at 2 µL/well, and incubation was performed at room temperature for 1 hour, and luminescence intensity was then measured for each well.
  • Analysis of cell growth inhibition rates (%) and effect of combined use under each of the conditions was performed in the same manner as in Example 14.
  • Synergy Scores for the combinations are shown in Table 4.
  • In the cell growth inhibition test for the HCC70 cell line, Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120. [Table 4]
    Synergy Scores for each combination in HCC70 cell line
    Erlotinib Poziotinib Mubritinib Brigatinib BIBF1120
    Compound (1) 0.93 0.95 3.41 4.11 1.65
    Antibody-drug conjugate (4) 1.38 0.64 1.44 1.60 1.30
  • Example 18: Cell growth inhibition test (5)
  • The human pancreatic cancer cell line BxPC-3 obtained from ATCC was used for evaluation. To a 1536-well cell culture plate, erlotinib (hydrochloride), poziotinib, mubritinib, brigatinib, or BIBF1120 prepared with DMSO to concentrations of 2 mM, 400 µM, 80 µM, 16 µM, 3.2 µM, and 640 nM or DMSO was added at 25 nL/well. Further, Compound (1) prepared with an RPMI1640 Medium containing 10% fetal bovine serum to concentrations of 80 nM, 32 nM, 13 nM, 5.1 nM, 2.0 nM, and 0.82 nM, or the antibody-drug conjugate (4) prepared to concentrations of 16 nM, 5.3 nM, 1.8 nM, 0.59 nM, 0.20 nM, and 0.066 nM was added at 2.5 µL/well. Subsequently, BxPC-3 cells suspended with an RPMI1640 Medium containing 10% fetal bovine serum to a concentration of 4 × 104 cells/mL were seeded at 2.5 µL/well, and cultured at 37°C under 5%CO2 for 6 days. After culturing, a solution obtained by diluting CellTiter-Glo 2.0 Assay (Promega Corporation) with an equivalent amount of Glo Lysis buffer, 1 × (Promega Corporation) was added at 2 µL/well, and incubation was performed at room temperature for 1 hour, and luminescence intensity was then measured for each well.
  • Analysis of cell growth inhibition rates (%) and effect of combined use under each of the conditions was performed in the same manner as in Example 14.
  • Synergy Scores for the combinations are shown in Table 5.
  • In the cell growth inhibition test for the BxPC-3 cell line, Compound (1) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120, and the antibody-drug conjugate (4) exhibited synergistic action when being combined with erlotinib (hydrochloride), poziotinib, brigatinib, or BIBF1120. [Table 5]
    Synergy Scores for each combination in BxPC-3 cell line
    Erlotinib Poziotinib Mubritinib Brigatinib BIBF1120
    Compound (1) 1.29 0.37 -0.40 0.93 0.28
    Antibody-drug conjugate (4) 1.84 0.39 -0.30 1.67 1.28
  • Example 19: Antitumor test (9)
  • MDA-MB-453 cells, a human breast cancer cell line, obtained from ATCC were suspended in Matrigel basement membrane matrix, and 1 × 107 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 11 days after the transplantation (Day 0). The antibody-drug conjugate (2) was diluted with ABS buffer, and intravenously administered to the tail vein of each mouse once a week at a dose of 1 mg/kg (a liquid volume of 10 mL/kg) for three weeks ( Days 0, 7, 14). Palbociclib was suspended in 0.5% methyl cellulose, and orally administered once a day, five times a week, at a dose of 50 mg/kg (a liquid volume of 10 mL/kg) for three weeks from Day 0. Single administration groups and a combined administration group with the antibody-drug conjugate (2) and palbociclib, and an ABS administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (2) and palbociclib are shown in Figure 21. TGI of single administration of palbociclib was 72%. The TGI of single administration of the antibody-drug conjugate (2) was 54%. For combined administration of the antibody-drug conjugate (2) and palbociclib, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (2) and single administration of palbociclib was found (P < 0.0001 and P = 0.004, respectively). The tumor growth inhibition (TGI: 93%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Example 20: Antitumor test (10)
  • MDA-MB-453 cells, a human breast cancer cell line, obtained from ATCC were suspended in Matrigel basement membrane matrix, and 1 × 107 cells were subcutaneously transplanted to the right flank of each female nude mouse, and the mice were randomly grouped 7 days after the transplantation (Day 0). The antibody-drug conjugate (2) was diluted with ABS buffer, and intravenously administered to the tail vein of each mouse once a week at a dose of 1 mg/kg (a liquid volume of 10 mL/kg) for three weeks ( Days 0, 7, 14). Alpelisib was suspended in 0.5% methyl cellulose, and orally administered once a day, five times a week, at a dose of 25 mg/kg (a liquid volume of 10 mL/kg) for three weeks from Day 0. Single administration groups and a combined administration group with the antibody-drug conjugate (2) and alpelisib, and an ABS administration group as a control group were established.
  • Results of combined use of the antibody-drug conjugate (2) and alpelisib are shown in Figure 22. TGI of single administration of alpelisib was 56%. The TGI of single administration of the antibody-drug conjugate (2) was 68%. For combined administration of the antibody-drug conjugate (2) and alpelisib, on the other hand, tumor growth inhibitory effect significantly superior to those of single administration of the antibody-drug conjugate (2) and single administration of alpelisib was found (P = 0.01 and P = 0.0001, respectively). The tumor growth inhibition (TGI: 86%) was higher than those of single administration of them, demonstrating strong effect of their combined use. No significant finding such as weight loss was observed in any of the single administration groups and combined administration group.
  • Free Text of Sequence Listing
    • SEQ ID NO: 1 - Amino acid sequence of a heavy chain of the anti-HER2 antibody
    • SEQ ID NO: 2 - Amino acid sequence of a light chain of the anti-HER2 antibody
    • SEQ ID NO: 3 - Amino acid sequence of a heavy chain of the anti-HER3 antibody
    • SEQ ID NO: 4 - Amino acid sequence of a light chain of the anti-HER3 antibody
    • SEQ ID NO: 5 - Amino acid sequence of a heavy chain of the anti-TROP2 antibody
    • SEQ ID NO: 6 - Amino acid sequence of a light chain of the anti-TROP2 antibody
    • SEQ ID NO: 7 - Amino acid sequence of a heavy chain of the anti-B7-H3 antibody
    • SEQ ID NO: 8 - Amino acid sequence of a light chain of the anti-B7-H3 antibody
    • SEQ ID NO: 9 - Amino acid sequence of a heavy chain of the anti-CDH6 antibody
    • SEQ ID NO: 10 - Amino acid sequence of a light chain of the anti-CDH6 antibody
    SEQUENCE LISTING
    • <110> DAIICHI SANKYO COMPANY, LIMITED
    • <120> COMBINATION OF ANTIBODY-DRUG CONJUGATE AND KINASE INHIBITOR
    • <130> DSFP1932WO
    • <150> JP2018-240049
      <151> 2018-12-21
    • <150> JP2019-085338
      <151> 2019-04-26
    • <160> 10
    • <170> PatentIn version 3.5
    • <210> 1
      <211> 450
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Heavy chain of anti-HER2 antibody
    • <400> 1
      Figure imgb0044
      Figure imgb0045
      Figure imgb0046
    • <210> 2
      <211> 214
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Light chain of anti-HER2 antibody
    • <400> 2
      Figure imgb0047
      Figure imgb0048
    • <210> 3
      <211> 447
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Heavy chain of anti-HER3 antibody
    • <400> 3
      Figure imgb0049
      Figure imgb0050
      Figure imgb0051
    • <210> 4
      <211> 220
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Light chain of anti-HER3 antibody
    • <400> 4
      Figure imgb0052
      Figure imgb0053
    • <210> 5
      <211> 470
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Heavy chain of anti-TROP2 antibody
    • <400> 5
      Figure imgb0054
      Figure imgb0055
      Figure imgb0056
    • <210> 6
      <211> 234
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Light chain of anti-TROP2 antibody
    • <400> 6
      Figure imgb0057
      Figure imgb0058
    • <210> 7
      <211> 471
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Heavy chain of anti-B7-H3 antibody
    • <400> 7
      Figure imgb0059
      Figure imgb0060
      Figure imgb0061
    • <210> 8
      <211> 233
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Light chain of anti-B7-H3 antibody
    • <400> 8
      Figure imgb0062
      Figure imgb0063
    • <210> 9
      <211> 471
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Heavy chain of anti-CDH6 antibody
    • <400> 9
      Figure imgb0064
      Figure imgb0065
      Figure imgb0066
    • <210> 10
      <211> 233
      <212> PRT
      <213> Artificial Sequence
    • <220>
    • <223> Light chain of anti-CDH6 antibody
    • <400> 10
      Figure imgb0067
      Figure imgb0068

Claims (124)

  1. A pharmaceutical composition wherein an antibody-drug conjugate and a kinase inhibitor are administered in combination, and the antibody-drug conjugate is an antibody-drug conjugate in which a drug-linker represented by the following formula:
    Figure imgb0069
    wherein A represents a connecting position to an antibody, is conjugated to the antibody via a thioether bond, and
    the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
  2. The pharmaceutical composition according to claim 1, wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
  3. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is a CDK4/6 inhibitor.
  4. The pharmaceutical composition according to claim 3, wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
  5. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an mTOR inhibitor.
  6. The pharmaceutical composition according to claim 5, wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
  7. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is a PI3K inhibitor.
  8. The pharmaceutical composition according to claim 7, wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
  9. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an RAF inhibitor.
  10. The pharmaceutical composition according to claim 9, wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
  11. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is a VEGFR inhibitor.
  12. The pharmaceutical composition according to claim 11, wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
  13. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is a KIT inhibitor.
  14. The pharmaceutical composition according to claim 13, wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
  15. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an RET inhibitor.
  16. The pharmaceutical composition according to claim 15, wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
  17. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is a PDGFR inhibitor.
  18. The pharmaceutical composition according to claim 17, wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
  19. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an FGFR inhibitor.
  20. The pharmaceutical composition according to claim 19, wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
  21. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an FLT3 inhibitor.
  22. The pharmaceutical composition according to claim 21, wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
  23. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an ALK inhibitor.
  24. The pharmaceutical composition according to claim 23, wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
  25. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is a CSF-1R inhibitor.
  26. The pharmaceutical composition according to claim 25, wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
  27. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an EGFR inhibitor.
  28. The pharmaceutical composition according to claim 27, wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
  29. The pharmaceutical composition according to claim 2, wherein the kinase inhibitor is an HER2 inhibitor.
  30. The pharmaceutical composition according to claim 29, wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
  31. The pharmaceutical composition according to any one of claims 1 to 30, wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
  32. The pharmaceutical composition according to claim 31, wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
  33. The pharmaceutical composition according to claim 32, wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
  34. The pharmaceutical composition according to claim 32, wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
  35. The pharmaceutical composition according to claim 32, wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
  36. The pharmaceutical composition according to claim 32, wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
  37. The pharmaceutical composition according to any one of claims 32 to 36, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
  38. The pharmaceutical composition according to claim 31, wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
  39. The pharmaceutical composition according to claim 38, wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
  40. The pharmaceutical composition according to claim 39, wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  41. The pharmaceutical composition according to any one of claims 38 to 40, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
  42. The pharmaceutical composition according to claim 31, wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
  43. The pharmaceutical composition according to claim 42, wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
  44. The pharmaceutical composition according to claim 43, wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  45. The pharmaceutical composition according to any one of claims 42 to 44, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
  46. The pharmaceutical composition according to claim 31, wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
  47. The pharmaceutical composition according to claim 46, wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
  48. The pharmaceutical composition according to claim 47, wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  49. The pharmaceutical composition according to any one of claims 46 to 48, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
  50. The pharmaceutical composition according to claim 31, wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
  51. The pharmaceutical composition according to claim 50, wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
  52. The pharmaceutical composition according to claim 51, wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  53. The pharmaceutical composition according to any one of claims 50 to 52, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
  54. The pharmaceutical composition according to any one of claims 1 to 53, wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
  55. The pharmaceutical composition according to any one of claims 1 to 54, wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
  56. The pharmaceutical composition according to claim 55, wherein the pharmaceutical composition is for use in treating breast cancer.
  57. The pharmaceutical composition according to claim 55, wherein the pharmaceutical composition is for use in treating colorectal cancer.
  58. The pharmaceutical composition according to claim 55, wherein the pharmaceutical composition is for use in treating gastric cancer.
  59. The pharmaceutical composition according to claim 55, wherein the pharmaceutical composition is for use in treating lung cancer.
  60. The pharmaceutical composition according to claim 55, wherein the pharmaceutical composition is for use in treating pancreatic cancer.
  61. The pharmaceutical composition according to claim 55, wherein the pharmaceutical composition is for use in treating kidney cancer.
  62. The pharmaceutical composition according to claim 55, wherein the pharmaceutical composition is for use in treating ovarian cancer.
  63. A pharmaceutical composition wherein an antibody-drug conjugate and a kinase inhibitor are administered in combination, and the antibody-drug conjugate is an antibody-drug conjugate represented by the following formula:
    Figure imgb0070
    Figure imgb0071
    wherein a drug-linker is conjugated to an antibody via a thioether bond, and n indicates the average number of units of the drug-linker conjugated per antibody molecule, and
    the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an AKT inhibitor, an ERK inhibitor, an MEK inhibitor, an RAF inhibitor, a CDK1 inhibitor, a CDK2 inhibitor, a CHK1 inhibitor, a WEE1 inhibitor, a PLK1 inhibitor, an Aurora kinase inhibitor, a Bcr-Abl inhibitor, an Src inhibitor, an EPH inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, a BTK inhibitor, an FLT3 inhibitor, an ALK inhibitor, a JAK inhibitor, an MET inhibitor, a CSF-1R inhibitor, an NTRK inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
  64. The pharmaceutical composition according to claim 63, wherein the kinase inhibitor is at least one selected from the group consisting of a CDK4/6 inhibitor, an mTOR inhibitor, a PI3K inhibitor, an RAF inhibitor, a VEGFR inhibitor, a KIT inhibitor, an RET inhibitor, a PDGFR inhibitor, an FGFR inhibitor, an FLT3 inhibitor, an ALK inhibitor, a CSF-1R inhibitor, an EGFR inhibitor, and an HER2 inhibitor.
  65. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is a CDK4/6 inhibitor.
  66. The pharmaceutical composition according to claim 65, wherein the CDK4/6 inhibitor is abemaciclib, palbociclib, ribociclib, trilaciclib, G1T38, PF-06873600, TP-1287, FN-1501, or KRX-0601, or a pharmacologically acceptable salt thereof.
  67. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an mTOR inhibitor.
  68. The pharmaceutical composition according to claim 67, wherein the mTOR inhibitor is everolimus, sirolimus, temsirolimus, TAK-228, CC-223, AZD8055, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, or PQR309, or a pharmacologically acceptable salt thereof.
  69. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is a PI3K inhibitor.
  70. The pharmaceutical composition according to claim 69, wherein the PI3K inhibitor is taselisib, alpelisib, TAK-117, GSK2636771, AZD8186, IPI-549, idelalisib, duvelisib, AMG319, buparlisib, pictilisib, pilaralisib, copanlisib, sonolisib, CH5132799, ZSTK474, GDC-0077, dactolisib, apitolisib, gedatolisib, LY3023414, PF-04691502, NVP-BGT226, PQR309, KRX-0601, or rigosertib, or a pharmacologically acceptable salt thereof.
  71. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an RAF inhibitor.
  72. The pharmaceutical composition according to claim 71, wherein the RAF inhibitor is regorafenib, sorafenib, vemurafenib, dabrafenib, encorafenib, RAF265, GDC-5573, LY3009120, or RO5126766, or a pharmacologically acceptable salt thereof.
  73. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is a VEGFR inhibitor.
  74. The pharmaceutical composition according to claim 73, wherein the VEGFR inhibitor is regorafenib, sorafenib, vandetanib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, cabozantinib, tivozanib, brivanib, linifanib, lucitanib, ilorasertib, or ENMD-2076, or a pharmacologically acceptable salt thereof.
  75. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is a KIT inhibitor.
  76. The pharmaceutical composition according to claim 75, wherein the KIT inhibitor is regorafenib, sorafenib, imatinib, ilorasertib, sunitinib, pazopanib, lenvatinib, or dasatinib, or a pharmacologically acceptable salt thereof.
  77. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an RET inhibitor.
  78. The pharmaceutical composition according to claim 77, wherein the RET inhibitor is regorafenib, sorafenib, vandetanib, lenvatinib, or sunitinib, or a pharmacologically acceptable salt thereof.
  79. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is a PDGFR inhibitor.
  80. The pharmaceutical composition according to claim 79, wherein the PDGFR inhibitor is regorafenib, sorafenib, sunitinib, axitinib, pazopanib, lenvatinib, nintedanib, ilorasertib, imatinib, nilotinib, or dasatinib, or a pharmacologically acceptable salt thereof.
  81. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an FGFR inhibitor.
  82. The pharmaceutical composition according to claim 81, wherein the FGFR inhibitor is regorafenib, sorafenib, lenvatinib, nintedanib, axitinib, or pazopanib, or a pharmacologically acceptable salt thereof.
  83. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an FLT3 inhibitor.
  84. The pharmaceutical composition according to claim 83, wherein the FLT3 inhibitor is gilteritinib, quizartinib, midostaurin, sorafenib, ilorasertib, ENMD-2076, or sunitinib, or a pharmacologically acceptable salt thereof.
  85. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an ALK inhibitor.
  86. The pharmaceutical composition according to claim 85, wherein the ALK inhibitor is brigatinib, crizotinib, ceritinib, alectinib, or lorlatinib, or a pharmacologically acceptable salt thereof.
  87. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is a CSF-1R inhibitor.
  88. The pharmaceutical composition according to claim 87, wherein the CSF-1R inhibitor is pexidartinib, BLZ-945, JNJ-40346527, JNJ-28312141, ilorasertib, imatinib, sunitinib, or axitinib, or a pharmacologically acceptable salt thereof.
  89. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an EGFR inhibitor.
  90. The pharmaceutical composition according to claim 89, wherein the EGFR inhibitor is gefitinib, erlotinib, afatinib, osimertinib, dacomitinib, lapatinib, neratinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
  91. The pharmaceutical composition according to claim 64, wherein the kinase inhibitor is an HER2 inhibitor.
  92. The pharmaceutical composition according to claim 91, wherein the HER2 inhibitor is tucatinib, neratinib, mubritinib, lapatinib, pyrotinib, or poziotinib, or a pharmacologically acceptable salt thereof.
  93. The pharmaceutical composition according to any one of claims 63 to 92, wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody, an anti-HER3 antibody, an anti-TROP2 antibody, an anti-B7-H3 antibody, or an anti-CDH6 antibody.
  94. The pharmaceutical composition according to claim 93, wherein the antibody in the antibody-drug conjugate is an anti-HER2 antibody.
  95. The pharmaceutical composition according to claim 94, wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising CDRH1 consisting of an amino acid sequence consisting of amino acid residues 26 to 33 of SEQ ID NO: 1, CDRH2 consisting of an amino acid sequence consisting of amino acid residues 51 to 58 of SEQ ID NO: 1, and CDRH3 consisting of an amino acid sequence consisting of amino acid residues 97 to 109 of SEQ ID NO: 1, and a light chain comprising CDRL1 consisting of an amino acid sequence consisting of amino acid residues 27 to 32 of SEQ ID NO: 2, CDRL2 consisting of an amino acid sequence consisting of amino acid residues 50 to 52 of SEQ ID NO: 2, and CDRL3 consisting of an amino acid sequence consisting of amino acid residues 89 to 97 of SEQ ID NO: 2.
  96. The pharmaceutical composition according to claim 94, wherein the anti-HER2 antibody is an antibody comprising a heavy chain comprising a heavy chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 120 of SEQ ID NO: 1 and a light chain comprising a light chain variable region consisting of an amino acid sequence consisting of amino acid residues 1 to 107 of SEQ ID NO: 2.
  97. The pharmaceutical composition according to claim 94, wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence represented by SEQ ID NO: 1 and a light chain consisting of an amino acid sequence represented by SEQ ID NO: 2.
  98. The pharmaceutical composition according to claim 94, wherein the anti-HER2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 1 to 449 of SEQ ID NO: 1 and a light chain consisting of an amino acid sequence consisting of amino acid residues 1 to 214 of SEQ ID NO: 2.
  99. The pharmaceutical composition according to any one of claims 94 to 98, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
  100. The pharmaceutical composition according to claim 93, wherein the antibody in the antibody-drug conjugate is an anti-HER3 antibody.
  101. The pharmaceutical composition according to claim 100, wherein the anti-HER3 antibody is an antibody comprising a heavy chain consisting of the amino acid sequence represented by SEQ ID NO: 3 and a light chain consisting of the amino acid sequence represented by SEQ ID NO: 4.
  102. The pharmaceutical composition according to claim 101, wherein the anti-HER3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  103. The pharmaceutical composition according to any one of claims 100 to 102, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
  104. The pharmaceutical composition according to claim 93, wherein the antibody in the antibody-drug conjugate is an anti-TROP2 antibody.
  105. The pharmaceutical composition according to claim 104, wherein the anti-TROP2 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 470 of SEQ ID NO: 5 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 234 of SEQ ID NO: 6.
  106. The pharmaceutical composition according to claim 105, wherein the anti-TROP2 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  107. The pharmaceutical composition according to any one of claims 104 to 106, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
  108. The pharmaceutical composition according to claim 93, wherein the antibody in the antibody-drug conjugate is an anti-B7-H3 antibody.
  109. The pharmaceutical composition according to claim 108, wherein the anti-B7-H3 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 7 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 8.
  110. The pharmaceutical composition according to claim 109, wherein the anti-B7-H3 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  111. The pharmaceutical composition according to any one of claims 108 to 110, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 3.5 to 4.5.
  112. The pharmaceutical composition according to claim 93, wherein the antibody in the antibody-drug conjugate is an anti-CDH6 antibody.
  113. The pharmaceutical composition according to claim 112, wherein the anti-CDH6 antibody is an antibody comprising a heavy chain consisting of an amino acid sequence consisting of amino acid residues 20 to 471 of SEQ ID NO: 9 and a light chain consisting of an amino acid sequence consisting of amino acid residues 21 to 233 of SEQ ID NO: 10.
  114. The pharmaceutical composition according to claim 113, wherein the anti-CDH6 antibody lacks a lysine residue at the carboxyl terminus of the heavy chain.
  115. The pharmaceutical composition according to any one of claims 112 to 114, wherein the average number of units of the drug-linker conjugated per antibody molecule in the antibody-drug conjugate is in the range of from 7 to 8.
  116. The pharmaceutical composition according to any one of claims 63 to 115, wherein the antibody-drug conjugate and the kinase inhibitor are separately contained as active components in different formulations, and are administered simultaneously or at different times.
  117. The pharmaceutical composition according to any one of claims 63 to 116, wherein the pharmaceutical composition is for use in treating at least one selected from the group consisting of breast cancer, gastric cancer, colorectal cancer, lung cancer, esophageal cancer, head-and-neck cancer, gastroesophageal junction adenocarcinoma, biliary tract cancer, Paget's disease, pancreatic cancer, ovarian cancer, uterine carcinosarcoma, urothelial cancer, prostate cancer, bladder cancer, gastrointestinal stromal tumor, gastrointestinal stromal tumor, uterine cervix cancer, squamous cell carcinoma, peritoneal cancer, liver cancer, hepatocellular cancer, endometrial cancer, kidney cancer, vulval cancer, thyroid cancer, penis cancer, leukemia, malignant lymphoma, plasmacytoma, myeloma, glioblastoma multiforme, osteosarcoma, sarcoma, and melanoma.
  118. The pharmaceutical composition according to claim 117, wherein the pharmaceutical composition is for use in treating breast cancer.
  119. The pharmaceutical composition according to claim 117, wherein the pharmaceutical composition is for use in treating colorectal cancer.
  120. The pharmaceutical composition according to claim 117, wherein the pharmaceutical composition is for use in treating gastric cancer.
  121. The pharmaceutical composition according to claim 117, wherein the pharmaceutical composition is for use in treating lung cancer.
  122. The pharmaceutical composition according to claim 117, wherein the pharmaceutical composition is for use in treating pancreatic cancer.
  123. The pharmaceutical composition according to claim 117, wherein the pharmaceutical composition is for use in treating kidney cancer.
  124. The pharmaceutical composition according to claim 117, wherein the pharmaceutical composition is for use in treating ovarian cancer.
EP19899188.7A 2018-12-21 2019-12-20 Combination of antibody-drug conjugate and kinase inhibitor Pending EP3903828A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2018240049 2018-12-21
JP2019085338 2019-04-26
PCT/JP2019/050017 WO2020130125A1 (en) 2018-12-21 2019-12-20 Combination of antibody-drug conjugate and kinase inhibitor

Publications (2)

Publication Number Publication Date
EP3903828A1 true EP3903828A1 (en) 2021-11-03
EP3903828A4 EP3903828A4 (en) 2022-10-05

Family

ID=71101395

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19899188.7A Pending EP3903828A4 (en) 2018-12-21 2019-12-20 Combination of antibody-drug conjugate and kinase inhibitor

Country Status (11)

Country Link
US (1) US20220040324A1 (en)
EP (1) EP3903828A4 (en)
JP (1) JPWO2020130125A1 (en)
KR (1) KR20210107069A (en)
CN (1) CN113195000A (en)
AU (1) AU2019407426A1 (en)
BR (1) BR112021011894A2 (en)
CA (1) CA3124330A1 (en)
SG (1) SG11202106635WA (en)
TW (1) TW202038957A (en)
WO (1) WO2020130125A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023228095A1 (en) * 2022-05-24 2023-11-30 Daiichi Sankyo Company, Limited Dosage regimen of an anti-cdh6 antibody-drug conjugate

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
CA2998469A1 (en) 2015-09-14 2017-03-23 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11419872B2 (en) * 2020-07-20 2022-08-23 Innoplexus Ag Phosphoinositide 3-kinase and Src inhibitors for treatment of pancreatic cancer
US11931424B2 (en) 2021-06-11 2024-03-19 Gilead Sciences, Inc. Combination MCL-1 inhibitors with anti-body drug conjugates
KR20240019283A (en) 2021-06-11 2024-02-14 길리애드 사이언시즈, 인코포레이티드 Combination use of MCL-1 inhibitor and anticancer drug
US11806405B1 (en) 2021-07-19 2023-11-07 Zeno Management, Inc. Immunoconjugates and methods
CN118201613A (en) * 2021-10-14 2024-06-14 江苏恒瑞医药股份有限公司 Use of anti-HER 2 antibody drug conjugate and tyrosine kinase inhibitor in combination for preparing tumor treatment drugs
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
CA3235986A1 (en) 2021-10-29 2023-05-04 Gilead Science, Inc. Cd73 compounds
WO2023091793A1 (en) * 2021-11-22 2023-05-25 Ohio State Innovation Foundation Methods and compositions for targeting alternative metabolism along with flt3 inhibitor-mediated antileukemic actions
AU2022419982A1 (en) 2021-12-22 2024-06-06 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
AU2022417491A1 (en) 2021-12-22 2024-05-23 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
US11945785B2 (en) 2021-12-30 2024-04-02 Biomea Fusion, Inc. Pyrazine compounds as inhibitors of FLT3
TW202340168A (en) 2022-01-28 2023-10-16 美商基利科學股份有限公司 Parp7 inhibitors
TW202346277A (en) 2022-03-17 2023-12-01 美商基利科學股份有限公司 Ikaros zinc finger family degraders and uses thereof
WO2023201268A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating tumor antigen expressing cancers
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
TW202400138A (en) 2022-04-21 2024-01-01 美商基利科學股份有限公司 Kras g12d modulating compounds
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds
WO2024097812A1 (en) 2022-11-04 2024-05-10 Gilead Sciences, Inc. Therapy for treating bladder cancer

Family Cites Families (114)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US713485A (en) 1902-05-15 1902-11-11 Joseph Neumeier Well-tube lifter.
JP2766360B2 (en) 1988-02-04 1998-06-18 協和醗酵工業株式会社 Staurosporine derivative
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
EP1400536A1 (en) 1991-06-14 2004-03-24 Genentech Inc. Method for making humanized antibodies
GB9221220D0 (en) 1992-10-09 1992-11-25 Sandoz Ag Organic componds
US5362718A (en) 1994-04-18 1994-11-08 American Home Products Corporation Rapamycin hydroxyesters
CA2216796C (en) 1995-03-30 2003-09-02 Pfizer Inc. Quinazoline derivatives
GB9508538D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
FR2741881B1 (en) 1995-12-01 1999-07-30 Centre Nat Rech Scient NOVEL PURINE DERIVATIVES HAVING IN PARTICULAR ANTI-PROLIFERATIVE PRORIETES AND THEIR BIOLOGICAL APPLICATIONS
US5989591A (en) 1997-03-14 1999-11-23 American Home Products Corporation Rapamycin formulations for oral administration
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
CO4940418A1 (en) 1997-07-18 2000-07-24 Novartis Ag MODIFICATION OF A CRYSTAL OF A DERIVATIVE OF N-PHENYL-2-PIRIMIDINAMINE, PROCESSES FOR ITS MANUFACTURE AND USE
PT1071700E (en) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
ME00275B (en) 1999-01-13 2011-02-10 Bayer Corp ?-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
ES2571230T3 (en) 1999-04-09 2016-05-24 Kyowa Hakko Kirin Co Ltd Procedure to control the activity of an immunofunctional molecule
MXPA01010292A (en) 1999-04-15 2002-10-23 Squibb Bristol Myers Co Cyclic protein tyrosine kinase inhibitors.
AU784045B2 (en) 1999-06-25 2006-01-19 Genentech Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
TWI262914B (en) 1999-07-02 2006-10-01 Agouron Pharma Compounds and pharmaceutical compositions for inhibiting protein kinases
US6762180B1 (en) 1999-10-13 2004-07-13 Boehringer Ingelheim Pharma Kg Substituted indolines which inhibit receptor tyrosine kinases
MXPA02004366A (en) 1999-11-05 2002-11-07 Astrazeneca Ab Quinazoline derivatives as vegf inhibitors.
CN1195755C (en) 1999-12-10 2005-04-06 辉瑞产品公司 Pyrrolo [2,3-d] pyrimidine compounds
MXPA02007005A (en) 2000-01-18 2004-09-06 Aventis Pharma Inc PSEUDOPOLYMORPH OF ()CIS2(2CHLOROPHENYL)5, 7DIHYDROXY8[4R(3SHYDROXY1METHYL)PIPERIDINYL]4Hminu s;1BENZOPYRAN4ONE.
SK287142B6 (en) 2000-02-15 2010-01-07 Sugen, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors, pharmacutical composition and use
GB0004888D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
PE20011178A1 (en) 2000-04-07 2001-11-19 Takeda Chemical Industries Ltd HETEROCYCLIC COMPOUNDS AND THEIR PRODUCTION
CN102311986B (en) 2000-10-06 2015-08-19 协和发酵麒麟株式会社 Produce the cell of antibody compositions
ES2282299T3 (en) 2000-10-20 2007-10-16 EISAI R&amp;D MANAGEMENT CO., LTD. AROMATIC COMPOUNDS WITH NITROGEN RINGS AS ANTICANCERIGEN AGENTS.
DE10063435A1 (en) 2000-12-20 2002-07-04 Boehringer Ingelheim Pharma Chinazoline derivatives, pharmaceuticals containing these compounds, their use and process for their preparation
HU230574B1 (en) 2000-12-21 2023-11-28 Novartis Ag Pyrimidineamines as angiogenesis modulators and pharmaceutical compositions containing them
US7235576B1 (en) 2001-01-12 2007-06-26 Bayer Pharmaceuticals Corporation Omega-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
PL209822B1 (en) 2001-04-27 2011-10-31 Kirin Pharma Kk Quinoline derivative having azolyl group and quinazoline derivative
WO2003024183A2 (en) 2001-09-14 2003-03-27 Arizona Board Of Regents On Behalf Of The University Of Arizona Wortmannin analogs and methods of using same
RU2337692C3 (en) 2001-10-30 2020-11-09 Новартис Аг STAUROSPORIN DERIVATIVES AS INHIBITORS OF FLT3 RECEPTOR TYROSINE KINASE ACTIVITY
DK1470124T3 (en) 2002-01-22 2006-04-18 Warner Lambert Co 2- (Pyridin-2-yl amino) pyrido [2,3] pyrimidin-7-one
US7235537B2 (en) 2002-03-13 2007-06-26 Array Biopharma, Inc. N3 alkylated benzimidazole derivatives as MEK inhibitors
ES2549159T3 (en) 2002-03-13 2015-10-23 Array Biopharma, Inc. N3-alkylated benzimidazole derivatives as MEK inhibitors
GB0215676D0 (en) 2002-07-05 2002-08-14 Novartis Ag Organic compounds
TWI329112B (en) 2002-07-19 2010-08-21 Bristol Myers Squibb Co Novel inhibitors of kinases
US7119200B2 (en) 2002-09-04 2006-10-10 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors
JP4790266B2 (en) 2002-09-30 2011-10-12 バイエル・シエリング・フアーマ・アクチエンゲゼルシヤフト Condensed azolepyrimidine derivatives
CA2537812C (en) 2003-09-26 2013-01-22 Exelixis, Inc. C-met modulators and method of use
DE10349423A1 (en) 2003-10-16 2005-06-16 Schering Ag Sulfoximine-substituted parimidines as CDK and / or VEGF inhibitors, their preparation and use as medicaments
PL1696920T3 (en) 2003-12-19 2015-03-31 Plexxikon Inc Compounds and methods for development of ret modulators
ME01267B (en) 2004-05-06 2013-06-20 Warner Lambert Co 4-phenylamino-quinazolin-6-yl-amides
JP4467616B2 (en) 2004-05-14 2010-05-26 ミレニアム・ファーマシューティカルズ・インコーポレイテッド Compounds and methods for blocking mitotic progression by inhibition of Aurora kinase
US7378423B2 (en) 2004-06-11 2008-05-27 Japan Tobacco Inc. Pyrimidine compound and medical use thereof
EP1773353B1 (en) 2004-07-19 2011-02-09 Onconova Therapeutics, Inc. Formulations for parenteral administration of (e)-2,6-dialkoxystryryl 4-substituted benzylsulfones
GEP20104906B (en) 2004-08-26 2010-02-25 Pfizer Enantiomerically pure aminoheteroaryl compounds as protein kinase inhibitors
DK1802579T3 (en) 2004-10-20 2014-01-20 Merck Serono Sa Derivatives of 3-arylaminopyridine
EP1827437B1 (en) * 2004-12-15 2011-11-02 Novartis AG Combinations of therapeutic agents for treating cancer
PL1912636T3 (en) 2005-07-21 2015-02-27 Ardea Biosciences Inc N-(arylamino)-sulfonamide inhibitors of mek
EA025871B9 (en) 2005-10-07 2017-08-31 Экселиксис, Инк. Mek inhibitors and methods of using the same
JP5270353B2 (en) 2005-10-07 2013-08-21 エクセリクシス, インク. Phosphatidylinositol 3-kinase inhibitor and method of use thereof
WO2007058628A1 (en) 2005-11-16 2007-05-24 S*Bio Pte Ltd Heteroalkyl linked pyrimidine derivatives
PT3184526T (en) 2005-12-13 2018-12-19 Incyte Holdings Corp Pyrrolo[2,3-d]pyrimidine derivatives as janus kinase inhibitor
PL1973545T3 (en) 2005-12-23 2013-05-31 Ariad Pharma Inc Bicyclic heteroaryl compounds
AR056857A1 (en) 2005-12-30 2007-10-24 U3 Pharma Ag DIRECTED ANTIBODIES TO HER-3 (RECEIVER OF THE HUMAN EPIDERMAL GROWTH FACTOR-3) AND ITS USES
JO2660B1 (en) 2006-01-20 2012-06-17 نوفارتيس ايه جي PI-3 Kinase inhibitors and methods of their use
EP2001358B1 (en) 2006-03-27 2016-07-13 University Of Maryland, Baltimore Glycoprotein synthesis and remodeling by enzymatic transglycosylation
MX2008013427A (en) 2006-04-19 2008-11-04 Novartis Ag 6-o-substituted benzoxazole and benzothiazole compounds and methods of inhibiting csf-1r signaling.
US7781433B2 (en) 2006-04-26 2010-08-24 Piramed Limited Pharmaceutical compounds
DK2046738T3 (en) 2006-06-21 2014-08-04 Piramal Entpr Ltd ENANTIOMERIC PURE FLAVONDERIVATIVES FOR THE TREATMENT OF PROLIFERATIVE DISORDERS, AND PROCESSES FOR THEIR PREPARATION
UA95641C2 (en) 2006-07-06 2011-08-25 Эррей Биофарма Инк. Hydroxylated cyclopenta [d] pyrimidines as akt protein kinase inhibitors
PT2526933E (en) 2006-09-22 2015-06-23 Pharmacyclics Inc Inhibitors of bruton's tyrosine kinase
ME02372B (en) 2006-11-22 2016-06-20 Incyte Holdings Corp Imidazotriazines and imidazopyrimidines as kinase inhibitors
MY180595A (en) 2006-12-07 2020-12-03 Genentech Inc Phosphoinositide 3-kinase inhibitor compounds and methods of use
UY30892A1 (en) 2007-02-07 2008-09-02 Smithkline Beckman Corp AKT ACTIVITY INHIBITORS
EP3248617A3 (en) 2007-02-16 2018-02-21 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
US8163923B2 (en) 2007-03-14 2012-04-24 Advenchen Laboratories, Llc Spiro substituted compounds as angiogenesis inhibitors
DE102007032507A1 (en) 2007-07-12 2009-04-02 Merck Patent Gmbh pyridazinone derivatives
PL2176231T3 (en) 2007-07-20 2017-04-28 Nerviano Medical Sciences S.R.L. Substituted indazole derivatives active as kinase inhibitors
JO3240B1 (en) 2007-10-17 2018-03-08 Janssen Pharmaceutica Nv Inhibitors of c-fms Kinase
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
TWI444382B (en) 2008-03-11 2014-07-11 Incyte Corp Azetidine and cyclobutane derivatives as jak inhibitors
UA103319C2 (en) 2008-05-06 2013-10-10 Глаксосмитклайн Ллк Thiazole- and oxazole-benzene sulfonamide compounds
LT2300013T (en) 2008-05-21 2017-12-27 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
CA2721692C (en) 2008-05-23 2014-01-14 Wyeth Llc Triazine compounds as p13 kinase and mtor inhibitors
UA104147C2 (en) 2008-09-10 2014-01-10 Новартис Аг Pyrrolidine dicarboxylic acid derivative and use thereof in the treatment of proliferative diseases
MY179042A (en) 2008-12-05 2020-10-26 Abbvie Bahamas Ltd Kinase inhibitors with improved cyp safety profile
JO2885B1 (en) 2008-12-22 2015-03-15 ايلي ليلي اند كومباني Protein kinase inhibitors
SG175707A1 (en) 2009-06-10 2011-12-29 Chugai Pharmaceutical Co Ltd Tetracyclic compound
JO3002B1 (en) 2009-08-28 2016-09-05 Irm Llc Compounds and compositions as protein kinase inhibitors
CN102020639A (en) 2009-09-14 2011-04-20 上海恒瑞医药有限公司 6-amido quinazoline or 3-cyano quinoline derivative, preparation method thereof and application of derivative to medicament
PL2578585T3 (en) 2010-05-31 2017-01-31 Ono Pharmaceutical Co., Ltd. Purinone derivative as btk kinase inhibitor
US8546566B2 (en) 2010-10-12 2013-10-01 Boehringer Ingelheim International Gmbh Process for manufacturing dihydropteridinones and intermediates thereof
KR101929593B1 (en) 2010-10-25 2018-12-14 쥐원 쎄라퓨틱스, 인크. Cdk inhibitors
AR083797A1 (en) 2010-11-10 2013-03-20 Novartis Ag DIMETHYL ACID SUCCINATE 7-CYCLOPENTIL-2- (5-PIPERAZIN-1-IL-PIRIDIN-2-IL-AMINO) -7H-PIRROLO- [2,3-D] PIRIMIDIN-6-CARBOXILICO, PROCESS FOR PREPARE IT, INTERMEDIARIES OF SUCH SYNTHESIS AND PREPARATION PROCESS OF THE SAME
MY192354A (en) 2011-07-19 2022-08-17 Merck Sharp & Dohme 4-imidazopyridazin-1-yl-benzamides and 4-imidazotriazin-1-yl-benzamides as btk-inhibitors
EP3686193B1 (en) 2011-07-27 2022-03-02 Astrazeneca AB 2-(2,4,5-substituted-anilino)pyrimidine compounds
SG11201401460PA (en) 2011-10-14 2014-09-26 Array Biopharma Inc POLYMORPHS OF ARRY-380, A SELECTIVE ErbB2 (HER2) INHIBITOR AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
PE20141404A1 (en) 2011-12-30 2014-10-28 Hanmi Pharm Ind Co Ltd DERIVATIVES OF HAVE [3,2-D] PYRIMIDINE WHICH HAVE INHIBITING ACTIVITY BY KINASES OF PROTEINS
IN2014DN06806A (en) 2012-02-10 2015-05-22 Univ Maryland
ES2848273T3 (en) 2012-03-05 2021-08-06 Gilead Calistoga Llc Polymorphic forms of (S) -2- (1- (9H-purin-6-ylamino) propyl) -5-fluoro-3-phenylquinazoline-4 (3H) -one
DK2822953T5 (en) 2012-03-06 2017-09-11 Pfizer Macrocyclic derivatives for the treatment of proliferative diseases
WO2014022975A1 (en) 2012-08-07 2014-02-13 上海创诺医药集团有限公司 N-(4-(3-amino-1h-indazol-4-yl)phenyl)-n'-(2-fluoro-5-methylphenyl)carbamide and preparation method for intermediate thereof
KR102584005B1 (en) 2012-10-11 2023-09-27 다이이찌 산쿄 가부시키가이샤 Method for producing a glycinamide compound
JP6272230B2 (en) 2012-10-19 2018-01-31 第一三共株式会社 Antibody-drug conjugate conjugated with a linker containing a hydrophilic structure
KR20140096571A (en) 2013-01-28 2014-08-06 한미약품 주식회사 Method for preparing 1-(4-(4-(3,4-dichloro-2-fluorophenylamino)-7-methoxyquinazolin-6-yloxy)piperidin-1-yl)prop-2-en-1-one
WO2014124230A2 (en) 2013-02-08 2014-08-14 Celgene Avilomics Research, Inc. Erk inhibitors and uses thereof
AU2014230812B2 (en) 2013-03-13 2016-04-07 F. Hoffmann-La Roche Ag Process for making benzoxazepin compounds
TWI731317B (en) 2013-12-10 2021-06-21 美商健臻公司 Tropomyosin-related kinase (trk) inhibitors
RU2743077C2 (en) 2013-12-25 2021-02-15 Дайити Санкио Компани, Лимитед Anti-trop2 antibody-drug conjugate
CN105829346B (en) 2014-01-31 2019-08-23 第一三共株式会社 Anti-HER 2-drug conjugates
JP2017114763A (en) 2014-03-26 2017-06-29 第一三共株式会社 Anti-CD98 antibody-drug conjugate
WO2015155976A1 (en) 2014-04-10 2015-10-15 第一三共株式会社 (anti-her2 antibody)-drug conjugate
ES2859648T3 (en) * 2014-04-10 2021-10-04 Daiichi Sankyo Co Ltd Antibody-anti-HER3 drug conjugate
TWI704151B (en) 2014-12-22 2020-09-11 美商美國禮來大藥廠 Erk inhibitors
ES2739749T3 (en) 2015-05-18 2020-02-03 Tolero Pharmaceuticals Inc Alvocidib prodrugs that have increased bioavailability
ES2938186T3 (en) 2015-06-29 2023-04-05 Daiichi Sankyo Co Ltd Method for the selective manufacture of an antibody-drug conjugate
TW202108592A (en) 2015-07-02 2021-03-01 瑞士商赫孚孟拉羅股份公司 Benzoxazepin oxazolidinone compounds and methods of use
HUE055978T2 (en) 2016-08-15 2022-01-28 Pfizer Pyridopyrimdinone cdk2/4/6 inhibitors
TW201834696A (en) * 2017-02-28 2018-10-01 學校法人近畿大學 Method for treating EGFR-TKI-resistant non-small cell lung cancer by administration of anti-HER3 antibody-drug conjugate
TW202330036A (en) 2017-05-15 2023-08-01 日商第一三共股份有限公司 Manufacturing method of antibody-drug conjugates
WO2019044947A1 (en) 2017-08-31 2019-03-07 第一三共株式会社 Improved method for producing antibody-drug conjugate

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023228095A1 (en) * 2022-05-24 2023-11-30 Daiichi Sankyo Company, Limited Dosage regimen of an anti-cdh6 antibody-drug conjugate

Also Published As

Publication number Publication date
US20220040324A1 (en) 2022-02-10
AU2019407426A1 (en) 2021-07-22
EP3903828A4 (en) 2022-10-05
WO2020130125A1 (en) 2020-06-25
SG11202106635WA (en) 2021-07-29
KR20210107069A (en) 2021-08-31
CN113195000A (en) 2021-07-30
BR112021011894A2 (en) 2021-09-08
CA3124330A1 (en) 2020-06-25
TW202038957A (en) 2020-11-01
JPWO2020130125A1 (en) 2021-11-04

Similar Documents

Publication Publication Date Title
EP3903828A1 (en) Combination of antibody-drug conjugate and kinase inhibitor
EP3834843A1 (en) Combination of antibody-drug conjugate and tubulin inhibitor
EP3909580A1 (en) Combination of antibody-drug conjugate with parp inhibitor
TW201834696A (en) Method for treating EGFR-TKI-resistant non-small cell lung cancer by administration of anti-HER3 antibody-drug conjugate
EP3831412A1 (en) Treatment of metastatic brain tumor by administration of antibody-drug conjugate
TW202024133A (en) Treatment for her3 mutated cancer comprising administering anti-her3 antibody-drug conjugate
US20230233540A1 (en) Combination of antibody-drug conjugate and cdk9 inhibitor
WO2021260583A1 (en) Combination of antibody-drug conjugate and dna-pk inhibitor
WO2021260582A1 (en) Combination of antibody-drug conjugate and aurora b inhibitor
EP4183420A1 (en) Combination of (anti-her2 antibody)-drug conjugate and her dimerization inhibitor
TW202216208A (en) Combination of antibody-drug conjugate and atr inhibitor
WO2023126822A1 (en) Combination of antibody-drug conjugate and rasg12c inhibitor

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20210624

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40054690

Country of ref document: HK

RAV Requested validation state of the european patent: fee paid

Extension state: TN

Effective date: 20210624

Extension state: MA

Effective date: 20210624

A4 Supplementary search report drawn up and despatched

Effective date: 20220901

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 16/32 20060101ALI20220826BHEP

Ipc: A61P 43/00 20060101ALI20220826BHEP

Ipc: A61P 35/02 20060101ALI20220826BHEP

Ipc: A61P 35/00 20060101ALI20220826BHEP

Ipc: A61K 45/06 20060101ALI20220826BHEP

Ipc: A61K 47/68 20170101AFI20220826BHEP

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230530